header logo image


Page 218«..1020..217218219220..230240..»

Autologous Stem Cell and Non Stem Based therapies Market Share, Size 2021 Global Industry Future Trends, Growth, Strategies,, Segmentation, In-depth…

February 1st, 2021 4:45 pm

Autologous Stem Cell and Non Stem Based therapies Market delivers a succinct analysis of industry size, regional growth and revenue forecasts for the upcoming years. The report further sheds light on significant challenges and the latest growth strategies adopted by manufacturers who are a part of the competitive spectrum of this business domain.

Autologous Stem Cell and Non Stem Based therapies Market: Global Size, Trends, Competitive, Historical & Forecast Analysis, 2021-2027. Rise in the prevalence of Cancer and Diabetes in all age groups population. Furthermore, the growing geriatric population is another key factor which drives the Autologous Stem Cell and Non Stem Based therapies Market.

Get Sample Copy of The Report@https://brandessenceresearch.com/requestSample/PostId/61

Scope Of Market Reports

Autologous Stem Cell transplantation is a process in which cells from which all blood cells develop are removed, preserved and later given to the same person after severe treatment. In autologous stem cell transplantation, the patient itself acts as stem cell donor. These cells are collected in advance while they are in remission and returned to the patient at a later stage i.e., after two months. They are used to replace stem cells which have been impaired by high doses of chemotherapy.It is important to realize that the processes required in a stem cell transplant are lengthy and complicated. A transplant involves a lot of preparation and a lot of care after procedure. Many people have a single autologous stem cell transplant while others mainly having myeloma or tumors; have two or more continuous transplants.

The initial step in an autologous stem cell transplant is gathering the stem cells. Physicians usually collect stem cells from the bloodstream (peripheral blood stem cells) in advance. A mobilization treatment is used. When the stem cells are in the bloodstream, then collection process starts.The blood is separated using an Apheresis machine. This procedure requires a few hours, and is repeated until the appropriate amount of stem cells is collected. Once the stem cells are harvested, they are frozen in our Stem Cell Processing and Cryopreservation Laboratory until its time to transplant.

Autologous Stem Cell and Non Stem Based therapies Market is segmented on the basis of Application, product, End user and Geography. Based upon ApplicationAutologous Stem Cell and Non Stem Based therapies Market is classified as Neurodegenerative Disorders,Autoimmune Diseases, cancer &Tumors, Cardiovascular Diseases and Others. Based on the ProductAutologous Stem Cell and Non Stem Based therapies Market is classified into Blood Pressure Monitoring Devices, Pulmonary Pressure Monitoring Devices and Intracranial Pressure Monitoring Devices. On the basis of End users Autologous Stem Cell and Non Stem Based therapies Market is classified into Hospitals, Ambulatory Surgical Centers and Others.

The regions covered in Autologous Stem Cell and Non Stem Based therapies Market report are North America, Europe, Asia-Pacific and Rest of the World. On the basis of country level, Global Melanoma Drug Market sub divided in to U.S., Mexico, Canada, U.K., France, Germany, Italy, China, Japan, India, South East Asia, GCC, Africa, etc.

Rising prevalence of cancer and diabetes among people across all age groups, growing geriatric population, increasing demand for autologous stem cell and non-stem cell based therapies is another factor, which is likely to create a heightened demand. Moreover, Favorable reimbursement policies across several nations are also boosting market. Risks and complications associated with the Autologous Stem Cell and Non Stem Based therapy such as diarrhea, hair loss, nausea, severe infections, vomiting, heart complications, and infertility and thehigh cost of autologous cellular therapies ranging from $500,000 to $1,000,000 restraint the market. Innovation of some newtherapies with improved efficacy, fewer side effects are expected to offer good opportunity for growth of Autologous Stem Cell and Non Stem Based therapies Market in the future.

Get Methodology of Report @https://brandessenceresearch.com/requestMethodology/PostId/61

North America is probable to attain the largest share of the Autologous Stem Cell and Non Stem Based therapies Market in terms of revenue and expected to hold the position followed by Europe region. This is due to less risk related with the treatment. Also, the demand for these treatments is high due to their ability to cure a significant number of infectious diseases. Autologous stem cell and non-stem cell based therapies do not require an outside donor hence the treatment is less infectious and cheap. However, Asia Pacific is expected to show the high growth in the forecast period. The demand in this region will be led by countries such as China, India, Malaysia, and Vietnam. The demand is likely to grow as autologous stem cell and non-stem cell based therapies aid in the efficient management of cardiovascular diseases as well. Rising healthcare facilities as well as increasing tax and reimbursement procedures is also estimated to help in the growth of the autologous stem cell and non-stem cell based therapies market in the Asia Pacific.

Furthermore, increase in awareness of disease and government initiatives for improving health care facilities are expected to boost the regional market to a certain extent.

By Application Analysis Neurodegenerative Disorders, Autoimmune Diseases, Cancer & Tumors, Cardiovascular Diseases, Others

By Product Analysis Blood Pressure Monitoring Devices, Pulmonary Pressure Monitoring Devices, Intracranial Pressure Monitoring Devices, Others

By End User Analysis Hospitals, Ambulatory Surgical centers, Others

North America, US, Mexico, Chily, Canada, Europe, UK, France, Germany, Italy, Asia Pacific, China, South Korea, Japan, India, Southeast Asia, Latin America, Brazil, The Middle East and Africa, GCC, Africa, Rest of Middle East and Africa

Get Full Report :@https://brandessenceresearch.com/chemical-materials/autologous-stem-cell-therapies-market

Related Report @https://www.prnewswire.com/news-releases/at-19-71cagr-zero-trust-security-market-size-is-projected-to-reach-94-35-billion-by-2027says-brandessence-market-research-301213906.html

https://www.marketwatch.com/press-release/at-1971-cagr-zero-trust-security-market-size-is-projected-to-reach-9435-billion-by-2027-says-brandessence-market-research-2021-01-25?tesla=y

https://www.marketwatch.com/press-release/at-cagr-of-1222-remote-patient-monitoring-market-size-is-expected-to-reach-usd-293638-million-by-2025-says-brandessence-market-research-2021-01-12?tesla=y

About Us:

Brandessence Market Research and Consulting Pvt. ltd.

Brandessence market research publishes market research reports & business insights produced by highly qualified and experienced industry analysts. Our research reports are available in a wide range of industry verticals including aviation, food & beverage, healthcare, ICT, Construction, Chemicals and lot more. Brand Essence Market Research report will be best fit for senior executives, business development managers, marketing managers, consultants, CEOs, CIOs, COOs, and Directors, governments, agencies, organizations and Ph.D. Students. We have a delivery center in Pune, India and our sales office is in London.

Contact us at: +44-2038074155 or mail us atsales@industrystatsreport.com

Website:https://brandessenceresearch.com/

Article:https://businessstatsnews.com

Blog:http://www.dailyindustrywatch.com

Blog:https://marketsize.biz

Blog:https://technologyindustrynews.com

Blog:https://marketstatsreport.com

Here is the original post:
Autologous Stem Cell and Non Stem Based therapies Market Share, Size 2021 Global Industry Future Trends, Growth, Strategies,, Segmentation, In-depth...

Read More...

Merck Receives Positive EU CHMP Opinion for Expanded Approval of KEYTRUDA (pembrolizumab) in Certain Patients With Relapsed or Refractory Classical…

February 1st, 2021 4:45 pm

KENILWORTH, N.J.--(BUSINESS WIRE)--Merck (NYSE: MRK), known as MSD outside the United States and Canada, today announced that the Committee for Medicinal Products for Human Use (CHMP) of the European Medicines Agency (EMA) has adopted a positive opinion recommending approval of an expanded label for KEYTRUDA, Mercks anti-PD-1 therapy. The opinion is recommending KEYTRUDA as monotherapy for the treatment of adult and pediatric patients aged 3 years and older with relapsed or refractory classical Hodgkin lymphoma (cHL) who have failed autologous stem cell transplant (ASCT) or following at least two prior therapies when ASCT is not a treatment option.

This recommendation is based on results from the pivotal Phase 3 KEYNOTE-204 trial, in which KEYTRUDA monotherapy demonstrated a significant improvement in progression-free survival (PFS) compared with brentuximab vedotin (BV), a commonly used treatment. KEYTRUDA reduced the risk of disease progression or death by 35% (HR=0.65 [95% CI, 0.48-0.88]; p=0.00271) and showed a median PFS of 13.2 months versus 8.3 months for patients treated with BV. The recommendation is also based on supportive data from an updated analysis of the KEYNOTE-087 trial, which supported the European Commissions (EC) approval of KEYTRUDA for the treatment of adult patients with relapsed or refractory cHL who have failed ASCT and BV or who are transplant ineligible and have failed BV. The CHMPs recommendation will now be reviewed by the EC for marketing authorization in the European Union (EU), and a final decision is expected in the first quarter of 2021. If approved, this will be the first pediatric indication for KEYTRUDA in the EU.

This positive opinion reinforces the importance of KEYTRUDA for certain adult and pediatric patients with relapsed or refractory classical Hodgkin lymphoma in the European Union, said Dr. Vicki Goodman, vice president, clinical research, Merck Research Laboratories. We look forward to the decision by the European Commission and will continue to expand our clinical development program in blood cancers with KEYTRUDA and our recently acquired investigational therapies to help address the unmet needs of patients.

Merck is studying KEYTRUDA across hematologic malignancies through a broad clinical program, including multiple registrational trials in cHL and primary mediastinal large B-cell lymphoma and more than 60 investigator-initiated studies across 15 tumors. In addition to KEYTRUDA, Merck is evaluating two clinical-stage assets for the treatment of patients with hematologic malignancies: MK-1026 (formerly ARQ 531), a Brutons tyrosine kinase inhibitor, and VLS-101, an antibody-drug conjugate targeting ROR1.

About KEYNOTE-204

KEYNOTE-204 (ClinicalTrials.gov, NCT02684292) is a randomized, open-label, Phase 3 trial evaluating KEYTRUDA monotherapy compared with BV for the treatment of patients with relapsed or refractory cHL. The primary endpoints are PFS and overall survival (OS), and the secondary endpoints include objective response rate (ORR), complete remission rate (CRR) and safety. The study enrolled 304 patients, aged 18 years and older, who were randomized to receive either:

About Hodgkin Lymphoma

Hodgkin lymphoma is a type of lymphoma that develops in the white blood cells called lymphocytes, which are part of the immune system. Hodgkin lymphoma can start almost anywhere most often in lymph nodes in the upper part of the body, with the most common sites being in the chest, neck or under the arms. Worldwide, there were approximately 83,000 new cases of Hodgkin lymphoma diagnosed, and more than 23,000 people died from the disease in 2020. In the EU, there were nearly 20,000 new cases of Hodgkin lymphoma diagnosed, and nearly 4,000 people died from the disease in 2020. Classical Hodgkin lymphoma accounts for more than nine in 10 cases of Hodgkin lymphoma in developed countries.

About KEYTRUDA (pembrolizumab) Injection, 100 mg

KEYTRUDA is an anti-PD-1 therapy that works by increasing the ability of the bodys immune system to help detect and fight tumor cells. KEYTRUDA is a humanized monoclonal antibody that blocks the interaction between PD-1 and its ligands, PD-L1 and PD-L2, thereby activating T lymphocytes which may affect both tumor cells and healthy cells.

Merck has the industrys largest immuno-oncology clinical research program. There are currently more than 1,300 trials studying KEYTRUDA across a wide variety of cancers and treatment settings. The KEYTRUDA clinical program seeks to understand the role of KEYTRUDA across cancers and the factors that may predict a patient's likelihood of benefitting from treatment with KEYTRUDA, including exploring several different biomarkers.

Selected KEYTRUDA (pembrolizumab) Indications in the U.S.

Melanoma

KEYTRUDA is indicated for the treatment of patients with unresectable or metastatic melanoma.

KEYTRUDA is indicated for the adjuvant treatment of patients with melanoma with involvement of lymph node(s) following complete resection.

Non-Small Cell Lung Cancer

KEYTRUDA, in combination with pemetrexed and platinum chemotherapy, is indicated for the first-line treatment of patients with metastatic nonsquamous non-small cell lung cancer (NSCLC), with no EGFR or ALK genomic tumor aberrations.

KEYTRUDA, in combination with carboplatin and either paclitaxel or paclitaxel protein-bound, is indicated for the first-line treatment of patients with metastatic squamous NSCLC.

KEYTRUDA, as a single agent, is indicated for the first-line treatment of patients with NSCLC expressing PD-L1 [tumor proportion score (TPS) 1%] as determined by an FDA-approved test, with no EGFR or ALK genomic tumor aberrations, and is stage III where patients are not candidates for surgical resection or definitive chemoradiation, or metastatic.

KEYTRUDA, as a single agent, is indicated for the treatment of patients with metastatic NSCLC whose tumors express PD-L1 (TPS 1%) as determined by an FDA-approved test, with disease progression on or after platinum-containing chemotherapy. Patients with EGFR or ALK genomic tumor aberrations should have disease progression on FDA-approved therapy for these aberrations prior to receiving KEYTRUDA.

Small Cell Lung Cancer

KEYTRUDA is indicated for the treatment of patients with metastatic small cell lung cancer (SCLC) with disease progression on or after platinum-based chemotherapy and at least 1 other prior line of therapy. This indication is approved under accelerated approval based on tumor response rate and durability of response. Continued approval for this indication may be contingent upon verification and description of clinical benefit in confirmatory trials.

Head and Neck Squamous Cell Cancer

KEYTRUDA, in combination with platinum and fluorouracil (FU), is indicated for the first-line treatment of patients with metastatic or with unresectable, recurrent head and neck squamous cell carcinoma (HNSCC).

KEYTRUDA, as a single agent, is indicated for the first-line treatment of patients with metastatic or with unresectable, recurrent HNSCC whose tumors express PD-L1 [combined positive score (CPS) 1] as determined by an FDA-approved test.

KEYTRUDA, as a single agent, is indicated for the treatment of patients with recurrent or metastatic HNSCC with disease progression on or after platinum-containing chemotherapy.

Classical Hodgkin Lymphoma

KEYTRUDA is indicated for the treatment of adult patients with relapsed or refractory classical Hodgkin lymphoma (cHL).

KEYTRUDA is indicated for the treatment of pediatric patients with refractory cHL, or cHL that has relapsed after 2 or more lines of therapy.

Primary Mediastinal Large B-Cell Lymphoma

KEYTRUDA is indicated for the treatment of adult and pediatric patients with refractory primary mediastinal large B-cell lymphoma (PMBCL), or who have relapsed after 2 or more prior lines of therapy. KEYTRUDA is not recommended for treatment of patients with PMBCL who require urgent cytoreductive therapy.

Urothelial Carcinoma

KEYTRUDA is indicated for the treatment of patients with locally advanced or metastatic urothelial carcinoma (mUC) who are not eligible for cisplatin-containing chemotherapy and whose tumors express PD-L1 (CPS 10), as determined by an FDA-approved test, or in patients who are not eligible for any platinum-containing chemotherapy regardless of PD-L1 status. This indication is approved under accelerated approval based on tumor response rate and duration of response. Continued approval for this indication may be contingent upon verification and description of clinical benefit in confirmatory trials.

KEYTRUDA is indicated for the treatment of patients with locally advanced or metastatic urothelial carcinoma (mUC) who have disease progression during or following platinum-containing chemotherapy or within 12 months of neoadjuvant or adjuvant treatment with platinum-containing chemotherapy.

KEYTRUDA is indicated for the treatment of patients with Bacillus Calmette-Guerin (BCG)-unresponsive, high-risk, non-muscle invasive bladder cancer (NMIBC) with carcinoma in situ (CIS) with or without papillary tumors who are ineligible for or have elected not to undergo cystectomy.

Microsatellite Instability-High or Mismatch Repair Deficient Cancer

KEYTRUDA is indicated for the treatment of adult and pediatric patients with unresectable or metastatic microsatellite instability-high (MSI-H) or mismatch repair deficient (dMMR)

This indication is approved under accelerated approval based on tumor response rate and durability of response. Continued approval for this indication may be contingent upon verification and description of clinical benefit in the confirmatory trials. The safety and effectiveness of KEYTRUDA in pediatric patients with MSI-H central nervous system cancers have not been established.

Microsatellite Instability-High or Mismatch Repair Deficient Colorectal Cancer

KEYTRUDA is indicated for the first-line treatment of patients with unresectable or metastatic MSI-H or dMMR colorectal cancer (CRC).

Gastric Cancer

KEYTRUDA is indicated for the treatment of patients with recurrent locally advanced or metastatic gastric or gastroesophageal junction (GEJ) adenocarcinoma whose tumors express PD-L1 (CPS 1) as determined by an FDA-approved test, with disease progression on or after two or more prior lines of therapy including fluoropyrimidine- and platinum-containing chemotherapy and if appropriate, HER2/neu-targeted therapy. This indication is approved under accelerated approval based on tumor response rate and durability of response. Continued approval for this indication may be contingent upon verification and description of clinical benefit in the confirmatory trials.

Esophageal Cancer

KEYTRUDA is indicated for the treatment of patients with recurrent locally advanced or metastatic squamous cell carcinoma of the esophagus whose tumors express PD-L1 (CPS 10) as determined by an FDA-approved test, with disease progression after one or more prior lines of systemic therapy.

Cervical Cancer

KEYTRUDA is indicated for the treatment of patients with recurrent or metastatic cervical cancer with disease progression on or after chemotherapy whose tumors express PD-L1 (CPS 1) as determined by an FDA-approved test. This indication is approved under accelerated approval based on tumor response rate and durability of response. Continued approval for this indication may be contingent upon verification and description of clinical benefit in the confirmatory trials.

Hepatocellular Carcinoma

KEYTRUDA is indicated for the treatment of patients with hepatocellular carcinoma (HCC) who have been previously treated with sorafenib. This indication is approved under accelerated approval based on tumor response rate and durability of response. Continued approval for this indication may be contingent upon verification and description of clinical benefit in the confirmatory trials.

Merkel Cell Carcinoma

KEYTRUDA is indicated for the treatment of adult and pediatric patients with recurrent locally advanced or metastatic Merkel cell carcinoma (MCC). This indication is approved under accelerated approval based on tumor response rate and durability of response. Continued approval for this indication may be contingent upon verification and description of clinical benefit in the confirmatory trials.

Renal Cell Carcinoma

KEYTRUDA, in combination with axitinib, is indicated for the first-line treatment of patients with advanced renal cell carcinoma (RCC).

Tumor Mutational Burden-High

KEYTRUDA is indicated for the treatment of adult and pediatric patients with unresectable or metastatic tumor mutational burden-high (TMB-H) [10 mutations/megabase] solid tumors, as determined by an FDA-approved test, that have progressed following prior treatment and who have no satisfactory alternative treatment options. This indication is approved under accelerated approval based on tumor response rate and durability of response. Continued approval for this indication may be contingent upon verification and description of clinical benefit in the confirmatory trials. The safety and effectiveness of KEYTRUDA in pediatric patients with TMB-H central nervous system cancers have not been established.

Cutaneous Squamous Cell Carcinoma

KEYTRUDA is indicated for the treatment of patients with recurrent or metastatic cutaneous squamous cell carcinoma (cSCC) that is not curable by surgery or radiation.

Triple-Negative Breast Cancer

KEYTRUDA, in combination with chemotherapy, is indicated for the treatment of patients with locally recurrent unresectable or metastatic triple-negative breast cancer (TNBC) whose tumors express PD-L1 (CPS 10) as determined by an FDA-approved test. This indication is approved under accelerated approval based on progression-free survival. Continued approval for this indication may be contingent upon verification and description of clinical benefit in the confirmatory trials.

Selected Important Safety Information for KEYTRUDA

Severe and Fatal Immune-Mediated Adverse Reactions

KEYTRUDA is a monoclonal antibody that belongs to a class of drugs that bind to either the programmed death receptor-1 (PD-1) or the programmed death ligand 1 (PD-L1), blocking the PD-1/PD-L1 pathway, thereby removing inhibition of the immune response, potentially breaking peripheral tolerance and inducing immune-mediated adverse reactions. Immune-mediated adverse reactions, which may be severe or fatal, can occur in any organ system or tissue, can affect more than one body system simultaneously, and can occur at any time after starting treatment or after discontinuation of treatment. Important immune-mediated adverse reactions listed here may not include all possible severe and fatal immune-mediated adverse reactions.

Monitor patients closely for symptoms and signs that may be clinical manifestations of underlying immune-mediated adverse reactions. Early identification and management are essential to ensure safe use of antiPD-1/PD-L1 treatments. Evaluate liver enzymes, creatinine, and thyroid function at baseline and periodically during treatment. In cases of suspected immune-mediated adverse reactions, initiate appropriate workup to exclude alternative etiologies, including infection. Institute medical management promptly, including specialty consultation as appropriate.

Withhold or permanently discontinue KEYTRUDA depending on severity of the immune-mediated adverse reaction. In general, if KEYTRUDA requires interruption or discontinuation, administer systemic corticosteroid therapy (1 to 2 mg/kg/day prednisone or equivalent) until improvement to Grade 1 or less. Upon improvement to Grade 1 or less, initiate corticosteroid taper and continue to taper over at least 1 month. Consider administration of other systemic immunosuppressants in patients whose adverse reactions are not controlled with corticosteroid therapy.

Immune-Mediated Pneumonitis

KEYTRUDA can cause immune-mediated pneumonitis. The incidence is higher in patients who have received prior thoracic radiation. Immune-mediated pneumonitis occurred in 3.4% (94/2799) of patients receiving KEYTRUDA, including fatal (0.1%), Grade 4 (0.3%), Grade 3 (0.9%), and Grade 2 (1.3%) reactions. Systemic corticosteroids were required in 67% (63/94) of patients. Pneumonitis led to permanent discontinuation of KEYTRUDA in 1.3% (36) and withholding in 0.9% (26) of patients. All patients who were withheld reinitiated KEYTRUDA after symptom improvement; of these, 23% had recurrence. Pneumonitis resolved in 59% of the 94 patients.

Pneumonitis occurred in 8% (31/389) of adult patients with cHL receiving KEYTRUDA as a single agent, including Grades 3-4 in 2.3% of patients. Patients received high-dose corticosteroids for a median duration of 10 days (range: 2 days to 53 months). Pneumonitis rates were similar in patients with and without prior thoracic radiation. Pneumonitis led to discontinuation of KEYTRUDA in 5.4% (21) of patients. Of the patients who developed pneumonitis, 42% of these patients interrupted KEYTRUDA, 68% discontinued KEYTRUDA, and 77% had resolution.

Immune-Mediated Colitis

KEYTRUDA can cause immune-mediated colitis, which may present with diarrhea. Cytomegalovirus infection/reactivation has been reported in patients with corticosteroid-refractory immune-mediated colitis. In cases of corticosteroid-refractory colitis, consider repeating infectious workup to exclude alternative etiologies. Immune-mediated colitis occurred in 1.7% (48/2799) of patients receiving KEYTRUDA, including Grade 4 (<0.1%), Grade 3 (1.1%), and Grade 2 (0.4%) reactions. Systemic corticosteroids were required in 69% (33/48); additional immunosuppressant therapy was required in 4.2% of patients. Colitis led to permanent discontinuation of KEYTRUDA in 0.5% (15) and withholding in 0.5% (13) of patients. All patients who were withheld reinitiated KEYTRUDA after symptom improvement; of these, 23% had recurrence. Colitis resolved in 85% of the 48 patients.

Hepatotoxicity and Immune-Mediated Hepatitis

KEYTRUDA as a Single Agent

KEYTRUDA can cause immune-mediated hepatitis. Immune-mediated hepatitis occurred in 0.7% (19/2799) of patients receiving KEYTRUDA, including Grade 4 (<0.1%), Grade 3 (0.4%), and Grade 2 (0.1%) reactions. Systemic corticosteroids were required in 68% (13/19) of patients; additional immunosuppressant therapy was required in 11% of patients. Hepatitis led to permanent discontinuation of KEYTRUDA in 0.2% (6) and withholding in 0.3% (9) of patients. All patients who were withheld reinitiated KEYTRUDA after symptom improvement; of these, none had recurrence. Hepatitis resolved in 79% of the 19 patients.

KEYTRUDA with Axitinib

KEYTRUDA in combination with axitinib can cause hepatic toxicity. Monitor liver enzymes before initiation of and periodically throughout treatment. Consider monitoring more frequently as compared to when the drugs are administered as single agents. For elevated liver enzymes, interrupt KEYTRUDA and axitinib, and consider administering corticosteroids as needed. With the combination of KEYTRUDA and axitinib, Grades 3 and 4 increased alanine aminotransferase (ALT) (20%) and increased aspartate aminotransferase (AST) (13%) were seen, which was at a higher frequency compared to KEYTRUDA alone. Fifty-nine percent of the patients with increased ALT received systemic corticosteroids. In patients with ALT 3 times upper limit of normal (ULN) (Grades 2-4, n=116), ALT resolved to Grades 0-1 in 94%. Among the 92 patients who were rechallenged with either KEYTRUDA (n=3) or axitinib (n=34) administered as a single agent or with both (n=55), recurrence of ALT 3 times ULN was observed in 1 patient receiving KEYTRUDA, 16 patients receiving axitinib, and 24 patients receiving both. All patients with a recurrence of ALT 3 ULN subsequently recovered from the event.

Immune-Mediated Endocrinopathies

Adrenal Insufficiency

KEYTRUDA can cause primary or secondary adrenal insufficiency. For Grade 2 or higher, initiate symptomatic treatment, including hormone replacement as clinically indicated. Withhold KEYTRUDA depending on severity. Adrenal insufficiency occurred in 0.8% (22/2799) of patients receiving KEYTRUDA, including Grade 4 (<0.1%), Grade 3 (0.3%), and Grade 2 (0.3%) reactions. Systemic corticosteroids were required in 77% (17/22) of patients; of these, the majority remained on systemic corticosteroids. Adrenal insufficiency led to permanent discontinuation of KEYTRUDA in <0.1% (1) and withholding in 0.3% (8) of patients. All patients who were withheld reinitiated KEYTRUDA after symptom improvement.

Hypophysitis

KEYTRUDA can cause immune-mediated hypophysitis. Hypophysitis can present with acute symptoms associated with mass effect such as headache, photophobia, or visual field defects. Hypophysitis can cause hypopituitarism. Initiate hormone replacement as indicated. Withhold or permanently discontinue KEYTRUDA depending on severity. Hypophysitis occurred in 0.6% (17/2799) of patients receiving KEYTRUDA, including Grade 4 (<0.1%), Grade 3 (0.3%), and Grade 2 (0.2%) reactions. Systemic corticosteroids were required in 94% (16/17) of patients; of these, the majority remained on systemic corticosteroids. Hypophysitis led to permanent discontinuation of KEYTRUDA in 0.1% (4) and withholding in 0.3% (7) of patients. All patients who were withheld reinitiated KEYTRUDA after symptom improvement.

Thyroid Disorders

KEYTRUDA can cause immune-mediated thyroid disorders. Thyroiditis can present with or without endocrinopathy. Hypothyroidism can follow hyperthyroidism. Initiate hormone replacement for hypothyroidism or institute medical management of hyperthyroidism as clinically indicated. Withhold or permanently discontinue KEYTRUDA depending on severity. Thyroiditis occurred in 0.6% (16/2799) of patients receiving KEYTRUDA, including Grade 2 (0.3%). None discontinued, but KEYTRUDA was withheld in <0.1% (1) of patients.

Hyperthyroidism occurred in 3.4% (96/2799) of patients receiving KEYTRUDA, including Grade 3 (0.1%) and Grade 2 (0.8%). It led to permanent discontinuation of KEYTRUDA in <0.1% (2) and withholding in 0.3% (7) of patients. All patients who were withheld reinitiated KEYTRUDA after symptom improvement. Hypothyroidism occurred in 8% (237/2799) of patients receiving KEYTRUDA, including Grade 3 (0.1%) and Grade 2 (6.2%). It led to permanent discontinuation of KEYTRUDA in <0.1% (1) and withholding in 0.5% (14) of patients. All patients who were withheld reinitiated KEYTRUDA after symptom improvement. The majority of patients with hypothyroidism required long-term thyroid hormone replacement. The incidence of new or worsening hypothyroidism was higher in 1185 patients with HNSCC, occurring in 16% of patients receiving KEYTRUDA as a single agent or in combination with platinum and FU, including Grade 3 (0.3%) hypothyroidism. The incidence of new or worsening hypothyroidism was higher in 389 adult patients with cHL (17%) receiving KEYTRUDA as a single agent, including Grade 1 (6.2%) and Grade 2 (10.8%) hypothyroidism.

Type 1 Diabetes Mellitus (DM), Which Can Present With Diabetic Ketoacidosis

Monitor patients for hyperglycemia or other signs and symptoms of diabetes. Initiate treatment with insulin as clinically indicated. Withhold KEYTRUDA depending on severity. Type 1 DM occurred in 0.2% (6/2799) of patients receiving KEYTRUDA. It led to permanent discontinuation in <0.1% (1) and withholding of KEYTRUDA in <0.1% (1). All patients who were withheld reinitiated KEYTRUDA after symptom improvement.

Immune-Mediated Nephritis With Renal Dysfunction

KEYTRUDA can cause immune-mediated nephritis. Immune-mediated nephritis occurred in 0.3% (9/2799) of patients receiving KEYTRUDA, including Grade 4 (<0.1%), Grade 3 (0.1%), and Grade 2 (0.1%) reactions. Systemic corticosteroids were required in 89% (8/9) of patients. Nephritis led to permanent discontinuation of KEYTRUDA in 0.1% (3) and withholding in 0.1% (3) of patients. All patients who were withheld reinitiated KEYTRUDA after symptom improvement; of these, none had recurrence. Nephritis resolved in 56% of the 9 patients.

Immune-Mediated Dermatologic Adverse Reactions

KEYTRUDA can cause immune-mediated rash or dermatitis. Exfoliative dermatitis, including Stevens-Johnson syndrome, drug rash with eosinophilia and systemic symptoms, and toxic epidermal necrolysis, has occurred with antiPD-1/PD-L1 treatments. Topical emollients and/or topical corticosteroids may be adequate to treat mild to moderate nonexfoliative rashes. Withhold or permanently discontinue KEYTRUDA depending on severity. Immune-mediated dermatologic adverse reactions occurred in 1.4% (38/2799) of patients receiving KEYTRUDA, including Grade 3 (1%) and Grade 2 (0.1%) reactions. Systemic corticosteroids were required in 40% (15/38) of patients. These reactions led to permanent discontinuation in 0.1% (2) and withholding of KEYTRUDA in 0.6% (16) of patients. All patients who were withheld reinitiated KEYTRUDA after symptom improvement; of these, 6% had recurrence. The reactions resolved in 79% of the 38 patients.

Other Immune-Mediated Adverse Reactions

The following clinically significant immune-mediated adverse reactions occurred at an incidence of <1% (unless otherwise noted) in patients who received KEYTRUDA or were reported with the use of other antiPD-1/PD-L1 treatments. Severe or fatal cases have been reported for some of these adverse reactions. Cardiac/Vascular: Myocarditis, pericarditis, vasculitis; Nervous System: Meningitis, encephalitis, myelitis and demyelination, myasthenic syndrome/myasthenia gravis (including exacerbation), Guillain-Barr syndrome, nerve paresis, autoimmune neuropathy; Ocular: Uveitis, iritis and other ocular inflammatory toxicities can occur. Some cases can be associated with retinal detachment. Various grades of visual impairment, including blindness, can occur. If uveitis occurs in combination with other immune-mediated adverse reactions, consider a Vogt-Koyanagi-Harada-like syndrome, as this may require treatment with systemic steroids to reduce the risk of permanent vision loss; Gastrointestinal: Pancreatitis, to include increases in serum amylase and lipase levels, gastritis, duodenitis; Musculoskeletal and Connective Tissue: Myositis/polymyositis rhabdomyolysis (and associated sequelae, including renal failure), arthritis (1.5%), polymyalgia rheumatica; Endocrine: Hypoparathyroidism; Hematologic/Immune: Hemolytic anemia, aplastic anemia, hemophagocytic lymphohistiocytosis, systemic inflammatory response syndrome, histiocytic necrotizing lymphadenitis (Kikuchi lymphadenitis), sarcoidosis, immune thrombocytopenic purpura, solid organ transplant rejection.

Infusion-Related Reactions

KEYTRUDA can cause severe or life-threatening infusion-related reactions, including hypersensitivity and anaphylaxis, which have been reported in 0.2% of 2799 patients receiving KEYTRUDA. Monitor for signs and symptoms of infusion-related reactions. Interrupt or slow the rate of infusion for Grade 1 or Grade 2 reactions. For Grade 3 or Grade 4 reactions, stop infusion and permanently discontinue KEYTRUDA.

Complications of Allogeneic Hematopoietic Stem Cell Transplantation (HSCT)

Fatal and other serious complications can occur in patients who receive allogeneic HSCT before or after antiPD-1/PD-L1 treatment. Transplant-related complications include hyperacute graft-versus-host disease (GVHD), acute and chronic GVHD, hepatic veno-occlusive disease after reduced intensity conditioning, and steroid-requiring febrile syndrome (without an identified infectious cause). These complications may occur despite intervening therapy between antiPD-1/PD-L1 treatment and allogeneic HSCT. Follow patients closely for evidence of these complications and intervene promptly. Consider the benefit vs risks of using antiPD-1/PD-L1 treatments prior to or after an allogeneic HSCT.

Increased Mortality in Patients With Multiple Myeloma

In trials in patients with multiple myeloma, the addition of KEYTRUDA to a thalidomide analogue plus dexamethasone resulted in increased mortality. Treatment of these patients with an antiPD-1/PD-L1 treatment in this combination is not recommended outside of controlled trials.

Embryofetal Toxicity

Based on its mechanism of action, KEYTRUDA can cause fetal harm when administered to a pregnant woman. Advise women of this potential risk. In females of reproductive potential, verify pregnancy status prior to initiating KEYTRUDA and advise them to use effective contraception during treatment and for 4 months after the last dose.

Adverse Reactions

In KEYNOTE-006, KEYTRUDA was discontinued due to adverse reactions in 9% of 555 patients with advanced melanoma; adverse reactions leading to permanent discontinuation in more than one patient were colitis (1.4%), autoimmune hepatitis (0.7%), allergic reaction (0.4%), polyneuropathy (0.4%), and cardiac failure (0.4%). The most common adverse reactions (20%) with KEYTRUDA were fatigue (28%), diarrhea (26%), rash (24%), and nausea (21%).

In KEYNOTE-054, KEYTRUDA was permanently discontinued due to adverse reactions in 14% of 509 patients; the most common (1%) were pneumonitis (1.4%), colitis (1.2%), and diarrhea (1%). Serious adverse reactions occurred in 25% of patients receiving KEYTRUDA. The most common adverse reaction (20%) with KEYTRUDA was diarrhea (28%).

Here is the original post:
Merck Receives Positive EU CHMP Opinion for Expanded Approval of KEYTRUDA (pembrolizumab) in Certain Patients With Relapsed or Refractory Classical...

Read More...

Merck Presents Results From Head-to-Head Phase 3 KEYNOTE-598 Trial Evaluating KEYTRUDA (pembrolizumab) in Combination With Ipilimumab Versus KEYTRUDA…

February 1st, 2021 4:45 pm

In KEYNOTE-598, the addition of ipilimumab to KEYTRUDA did not improve overall survival or progression-free survival, and patients who received the combination were more likely to experience serious side effects than those who received KEYTRUDA monotherapy, said Dr. Michael Boyer, chief clinical officer and conjoint chair of thoracic oncology, Chris OBrien Lifehouse, Camperdown, NSW, Australia. KEYTRUDA monotherapy remains a standard of care for the first-line treatment of certain patients with metastatic non-small cell lung cancer whose tumors express PD-L1.

As a leader in lung cancer, we are pursuing a broad clinical program to better understand the potential of KEYTRUDA-based combinations to improve survival outcomes for patients with this devastating disease, said Dr. Roy Baynes, senior vice president and head of global clinical development, chief medical officer, Merck Research Laboratories. KEYNOTE-598 is the first head-to-head study designed to answer the question of whether combining KEYTRUDA with ipilimumab provided additional clinical benefits beyond treatment with KEYTRUDA alone in certain patients with metastatic non-small cell lung cancer. The results are clear the combination did not add clinical benefit but did add toxicity.

These results were presented in the Presidential Symposium at the IASLC 2020 World Conference on Lung Cancer hosted by the International Association for the Study of Lung Cancer on Friday, Jan. 29 and published in the Journal of Clinical Oncology. As previously announced in Nov. 2020, the study was discontinued due to futility based on the recommendation of an independent Data Monitoring Committee (DMC), which determined the benefit/risk profile of KEYTRUDA in combination with ipilimumab did not support continuing the trial. The DMC also advised that patients in the study discontinue treatment with ipilimumab/placebo.

KEYNOTE-598 Study Design and Additional Data (Late-Breaking Abstract #PS01.09)

KEYNOTE-598 (ClinicalTrials.gov, NCT03302234) is a randomized, double-blind, Phase 3 trial designed to evaluate KEYTRUDA in combination with ipilimumab compared to KEYTRUDA monotherapy as first-line treatment for patients with metastatic NSCLC without EGFR or ALK genomic tumor aberrations and whose tumors express PD-L1 (TPS 50%). The dual primary endpoints are OS and PFS. Secondary endpoints include objective response rate (ORR), duration of response (DOR) and safety.

The study enrolled 568 patients who were randomized 1:1 to receive KEYTRUDA (200 mg intravenously [IV] on Day 1 of each three-week cycle for up to 35 cycles) in combination with ipilimumab (1 mg/kg IV on Day 1 of each six-week cycle for up to 18 cycles); or KEYTRUDA (200 mg IV on Day 1 of each three-week cycle for up to 35 cycles) as monotherapy. Non-binding futility criteria for the study were based on restricted mean survival time (RMST), an alternative outcome measure estimated as the area under the survival curve through a fixed timepoint. The pre-specified criteria were differences in RMST for KEYTRUDA in combination with ipilimumab and KEYTRUDA monotherapy of 0.2 at the maximum observation time and 0.1 at 24 months of follow-up.

As of data cut-off, the median study follow-up was 20.6 months. Findings showed the median OS was 21.4 months for patients randomized to KEYTRUDA in combination with ipilimumab (n=284) versus 21.9 months for those randomized to KEYTRUDA monotherapy (n=284) (HR=1.08 [95% CI, 0.85-1.37]; p=0.74). The differences in RMST for KEYTRUDA in combination with ipilimumab and KEYTRUDA monotherapy were -0.56 at the maximum observation time and -0.52 at 24 months, meeting the futility criteria for the trial and confirming the benefit/risk profile of the combination did not support continuing the study. Additionally, the median PFS was 8.2 months for patients randomized to KEYTRUDA in combination with ipilimumab versus 8.4 months for those randomized to KEYTRUDA monotherapy (HR=1.06 [95% CI, 0.86-1.30]; p=0.72). In both arms of the study, ORR was 45.4%; the median DOR was 16.1 months for patients randomized to KEYTRUDA in combination with ipilimumab versus 17.3 months for those randomized to KEYTRUDA monotherapy.

No new safety signals for KEYTRUDA monotherapy were observed. Treatment-related adverse events (TRAEs) occurred in 76.2% of patients treated with KEYTRUDA in combination with ipilimumab versus 68.3% of patients treated with KEYTRUDA monotherapy. Of these TRAEs, 35.1% vs. 19.6% were Grade 3-5, 27.7% vs. 13.9% were serious, 6.0% vs. 3.2% led to discontinuation of ipilimumab or placebo, 19.1% vs. 7.5% led to discontinuation of both drugs and 2.5% vs. 0.0% (no patients) led to death. Additionally, immune-mediated adverse events (AEs) and infusion reactions occurred in 44.7% of patients treated with KEYTRUDA in combination with ipilimumab versus 32.4% of patients treated with KEYTRUDA monotherapy. Of these immune-mediated AEs, 20.2% vs. 7.8% were Grade 3-5, 19.1% vs. 7.1% were serious, 1.8% vs. 1.1% led to discontinuation of ipilimumab or placebo, 12.1% vs. 4.3% led to discontinuation of both drugs and 2.1% vs. 0.0% (no patients) led to death.

About Lung Cancer

Lung cancer, which forms in the tissues of the lungs, usually within cells lining the air passages, is the leading cause of cancer death worldwide. Each year, more people die of lung cancer than die of colon and breast cancers combined. The two main types of lung cancer are non-small cell and small cell. Non-small cell lung cancer (NSCLC) is the most common type of lung cancer, accounting for about 85% of all cases. Small cell lung cancer (SCLC) accounts for about 10% to 15% of all lung cancers. Before 2014, the five-year survival rate for patients diagnosed in the U.S. with NSCLC and SCLC was estimated to be 5% and 6%, respectively.

About KEYTRUDA (pembrolizumab) Injection, 100 mg

KEYTRUDA is an anti-PD-1 therapy that works by increasing the ability of the bodys immune system to help detect and fight tumor cells. KEYTRUDA is a humanized monoclonal antibody that blocks the interaction between PD-1 and its ligands, PD-L1 and PD-L2, thereby activating T lymphocytes which may affect both tumor cells and healthy cells.

Merck has the industrys largest immuno-oncology clinical research program. There are currently more than 1,300 trials studying KEYTRUDA across a wide variety of cancers and treatment settings. The KEYTRUDA clinical program seeks to understand the role of KEYTRUDA across cancers and the factors that may predict a patient's likelihood of benefitting from treatment with KEYTRUDA, including exploring several different biomarkers.

Selected KEYTRUDA (pembrolizumab) Indications in the U.S.

Melanoma

KEYTRUDA is indicated for the treatment of patients with unresectable or metastatic melanoma.

KEYTRUDA is indicated for the adjuvant treatment of patients with melanoma with involvement of lymph node(s) following complete resection.

Non-Small Cell Lung Cancer

KEYTRUDA, in combination with pemetrexed and platinum chemotherapy, is indicated for the first-line treatment of patients with metastatic nonsquamous non-small cell lung cancer (NSCLC), with no EGFR or ALK genomic tumor aberrations.

KEYTRUDA, in combination with carboplatin and either paclitaxel or paclitaxel protein-bound, is indicated for the first-line treatment of patients with metastatic squamous NSCLC.

KEYTRUDA, as a single agent, is indicated for the first-line treatment of patients with NSCLC expressing PD-L1 [tumor proportion score (TPS) 1%] as determined by an FDA-approved test, with no EGFR or ALK genomic tumor aberrations, and is stage III where patients are not candidates for surgical resection or definitive chemoradiation, or metastatic.

KEYTRUDA, as a single agent, is indicated for the treatment of patients with metastatic NSCLC whose tumors express PD-L1 (TPS 1%) as determined by an FDA-approved test, with disease progression on or after platinum-containing chemotherapy. Patients with EGFR or ALK genomic tumor aberrations should have disease progression on FDA-approved therapy for these aberrations prior to receiving KEYTRUDA.

Small Cell Lung Cancer

KEYTRUDA is indicated for the treatment of patients with metastatic small cell lung cancer (SCLC) with disease progression on or after platinum-based chemotherapy and at least 1 other prior line of therapy. This indication is approved under accelerated approval based on tumor response rate and durability of response. Continued approval for this indication may be contingent upon verification and description of clinical benefit in confirmatory trials.

Head and Neck Squamous Cell Cancer

KEYTRUDA, in combination with platinum and fluorouracil (FU), is indicated for the first-line treatment of patients with metastatic or with unresectable, recurrent head and neck squamous cell carcinoma (HNSCC).

KEYTRUDA, as a single agent, is indicated for the first-line treatment of patients with metastatic or with unresectable, recurrent HNSCC whose tumors express PD-L1 [combined positive score (CPS) 1] as determined by an FDA-approved test.

KEYTRUDA, as a single agent, is indicated for the treatment of patients with recurrent or metastatic HNSCC with disease progression on or after platinum-containing chemotherapy.

Classical Hodgkin Lymphoma

KEYTRUDA is indicated for the treatment of adult patients with relapsed or refractory classical Hodgkin lymphoma (cHL).

KEYTRUDA is indicated for the treatment of pediatric patients with refractory cHL, or cHL that has relapsed after 2 or more lines of therapy.

Primary Mediastinal Large B-Cell Lymphoma

KEYTRUDA is indicated for the treatment of adult and pediatric patients with refractory primary mediastinal large B-cell lymphoma (PMBCL), or who have relapsed after 2 or more prior lines of therapy. KEYTRUDA is not recommended for treatment of patients with PMBCL who require urgent cytoreductive therapy.

Urothelial Carcinoma

KEYTRUDA is indicated for the treatment of patients with locally advanced or metastatic urothelial carcinoma (mUC) who are not eligible for cisplatin-containing chemotherapy and whose tumors express PD-L1 (CPS 10), as determined by an FDA-approved test, or in patients who are not eligible for any platinum-containing chemotherapy regardless of PD-L1 status. This indication is approved under accelerated approval based on tumor response rate and duration of response. Continued approval for this indication may be contingent upon verification and description of clinical benefit in confirmatory trials.

KEYTRUDA is indicated for the treatment of patients with locally advanced or metastatic urothelial carcinoma (mUC) who have disease progression during or following platinum-containing chemotherapy or within 12 months of neoadjuvant or adjuvant treatment with platinum-containing chemotherapy.

KEYTRUDA is indicated for the treatment of patients with Bacillus Calmette-Guerin (BCG)-unresponsive, high-risk, non-muscle invasive bladder cancer (NMIBC) with carcinoma in situ (CIS) with or without papillary tumors who are ineligible for or have elected not to undergo cystectomy.

Microsatellite Instability-High or Mismatch Repair Deficient Cancer

KEYTRUDA is indicated for the treatment of adult and pediatric patients with unresectable or metastatic microsatellite instability-high (MSI-H) or mismatch repair deficient (dMMR)

This indication is approved under accelerated approval based on tumor response rate and durability of response. Continued approval for this indication may be contingent upon verification and description of clinical benefit in the confirmatory trials. The safety and effectiveness of KEYTRUDA in pediatric patients with MSI-H central nervous system cancers have not been established.

Microsatellite Instability-High or Mismatch Repair Deficient Colorectal Cancer

KEYTRUDA is indicated for the first-line treatment of patients with unresectable or metastatic MSI-H or dMMR colorectal cancer (CRC).

Gastric Cancer

KEYTRUDA is indicated for the treatment of patients with recurrent locally advanced or metastatic gastric or gastroesophageal junction (GEJ) adenocarcinoma whose tumors express PD-L1 (CPS 1) as determined by an FDA-approved test, with disease progression on or after two or more prior lines of therapy including fluoropyrimidine- and platinum-containing chemotherapy and if appropriate, HER2/neu-targeted therapy. This indication is approved under accelerated approval based on tumor response rate and durability of response. Continued approval for this indication may be contingent upon verification and description of clinical benefit in the confirmatory trials.

Esophageal Cancer

KEYTRUDA is indicated for the treatment of patients with recurrent locally advanced or metastatic squamous cell carcinoma of the esophagus whose tumors express PD-L1 (CPS 10) as determined by an FDA-approved test, with disease progression after one or more prior lines of systemic therapy.

Cervical Cancer

KEYTRUDA is indicated for the treatment of patients with recurrent or metastatic cervical cancer with disease progression on or after chemotherapy whose tumors express PD-L1 (CPS 1) as determined by an FDA-approved test. This indication is approved under accelerated approval based on tumor response rate and durability of response. Continued approval for this indication may be contingent upon verification and description of clinical benefit in the confirmatory trials.

Hepatocellular Carcinoma

KEYTRUDA is indicated for the treatment of patients with hepatocellular carcinoma (HCC) who have been previously treated with sorafenib. This indication is approved under accelerated approval based on tumor response rate and durability of response. Continued approval for this indication may be contingent upon verification and description of clinical benefit in the confirmatory trials.

Merkel Cell Carcinoma

KEYTRUDA is indicated for the treatment of adult and pediatric patients with recurrent locally advanced or metastatic Merkel cell carcinoma (MCC). This indication is approved under accelerated approval based on tumor response rate and durability of response. Continued approval for this indication may be contingent upon verification and description of clinical benefit in the confirmatory trials.

Renal Cell Carcinoma

KEYTRUDA, in combination with axitinib, is indicated for the first-line treatment of patients with advanced renal cell carcinoma (RCC).

Tumor Mutational Burden-High

KEYTRUDA is indicated for the treatment of adult and pediatric patients with unresectable or metastatic tumor mutational burden-high (TMB-H) [10 mutations/megabase] solid tumors, as determined by an FDA-approved test, that have progressed following prior treatment and who have no satisfactory alternative treatment options. This indication is approved under accelerated approval based on tumor response rate and durability of response. Continued approval for this indication may be contingent upon verification and description of clinical benefit in the confirmatory trials. The safety and effectiveness of KEYTRUDA in pediatric patients with TMB-H central nervous system cancers have not been established.

Cutaneous Squamous Cell Carcinoma

KEYTRUDA is indicated for the treatment of patients with recurrent or metastatic cutaneous squamous cell carcinoma (cSCC) that is not curable by surgery or radiation.

Triple-Negative Breast Cancer

KEYTRUDA, in combination with chemotherapy, is indicated for the treatment of patients with locally recurrent unresectable or metastatic triple-negative breast cancer (TNBC) whose tumors express PD-L1 (CPS 10) as determined by an FDA-approved test.

This indication is approved under accelerated approval based on progression-free survival. Continued approval for this indication may be contingent upon verification and description of clinical benefit in the confirmatory trials.

Selected Important Safety Information for KEYTRUDA

Severe and Fatal Immune-Mediated Adverse Reactions

KEYTRUDA is a monoclonal antibody that belongs to a class of drugs that bind to either the programmed death receptor-1 (PD-1) or the programmed death ligand 1 (PD-L1), blocking the PD-1/PD-L1 pathway, thereby removing inhibition of the immune response, potentially breaking peripheral tolerance and inducing immune-mediated adverse reactions. Immune-mediated adverse reactions, which may be severe or fatal, can occur in any organ system or tissue, can affect more than one body system simultaneously, and can occur at any time after starting treatment or after discontinuation of treatment. Important immune-mediated adverse reactions listed here may not include all possible severe and fatal immune-mediated adverse reactions.

Monitor patients closely for symptoms and signs that may be clinical manifestations of underlying immune-mediated adverse reactions. Early identification and management are essential to ensure safe use of antiPD-1/PD-L1 treatments. Evaluate liver enzymes, creatinine, and thyroid function at baseline and periodically during treatment. In cases of suspected immune-mediated adverse reactions, initiate appropriate workup to exclude alternative etiologies, including infection. Institute medical management promptly, including specialty consultation as appropriate.

Withhold or permanently discontinue KEYTRUDA depending on severity of the immune-mediated adverse reaction. In general, if KEYTRUDA requires interruption or discontinuation, administer systemic corticosteroid therapy (1 to 2 mg/kg/day prednisone or equivalent) until improvement to Grade 1 or less. Upon improvement to Grade 1 or less, initiate corticosteroid taper and continue to taper over at least 1 month. Consider administration of other systemic immunosuppressants in patients whose adverse reactions are not controlled with corticosteroid therapy.

Immune-Mediated Pneumonitis

KEYTRUDA can cause immune-mediated pneumonitis. The incidence is higher in patients who have received prior thoracic radiation. Immune-mediated pneumonitis occurred in 3.4% (94/2799) of patients receiving KEYTRUDA, including fatal (0.1%), Grade 4 (0.3%), Grade 3 (0.9%), and Grade 2 (1.3%) reactions. Systemic corticosteroids were required in 67% (63/94) of patients. Pneumonitis led to permanent discontinuation of KEYTRUDA in 1.3% (36) and withholding in 0.9% (26) of patients. All patients who were withheld reinitiated KEYTRUDA after symptom improvement; of these, 23% had recurrence. Pneumonitis resolved in 59% of the 94 patients.

Pneumonitis occurred in 8% (31/389) of adult patients with cHL receiving KEYTRUDA as a single agent, including Grades 3-4 in 2.3% of patients. Patients received high-dose corticosteroids for a median duration of 10 days (range: 2 days to 53 months). Pneumonitis rates were similar in patients with and without prior thoracic radiation. Pneumonitis led to discontinuation of KEYTRUDA in 5.4% (21) of patients. Of the patients who developed pneumonitis, 42% interrupted KEYTRUDA, 68% discontinued KEYTRUDA, and 77% had resolution.

Immune-Mediated Colitis

KEYTRUDA can cause immune-mediated colitis, which may present with diarrhea. Cytomegalovirus infection/reactivation has been reported in patients with corticosteroid-refractory immune-mediated colitis. In cases of corticosteroid-refractory colitis, consider repeating infectious workup to exclude alternative etiologies. Immune-mediated colitis occurred in 1.7% (48/2799) of patients receiving KEYTRUDA, including Grade 4 (<0.1%), Grade 3 (1.1%), and Grade 2 (0.4%) reactions. Systemic corticosteroids were required in 69% (33/48); additional immunosuppressant therapy was required in 4.2% of patients. Colitis led to permanent discontinuation of KEYTRUDA in 0.5% (15) and withholding in 0.5% (13) of patients. All patients who were withheld reinitiated KEYTRUDA after symptom improvement; of these, 23% had recurrence. Colitis resolved in 85% of the 48 patients.

Hepatotoxicity and Immune-Mediated Hepatitis

KEYTRUDA as a Single Agent

KEYTRUDA can cause immune-mediated hepatitis. Immune-mediated hepatitis occurred in 0.7% (19/2799) of patients receiving KEYTRUDA, including Grade 4 (<0.1%), Grade 3 (0.4%), and Grade 2 (0.1%) reactions. Systemic corticosteroids were required in 68% (13/19) of patients; additional immunosuppressant therapy was required in 11% of patients. Hepatitis led to permanent discontinuation of KEYTRUDA in 0.2% (6) and withholding in 0.3% (9) of patients. All patients who were withheld reinitiated KEYTRUDA after symptom improvement; of these, none had recurrence. Hepatitis resolved in 79% of the 19 patients.

KEYTRUDA with Axitinib

KEYTRUDA in combination with axitinib can cause hepatic toxicity. Monitor liver enzymes before initiation of and periodically throughout treatment. Consider monitoring more frequently as compared to when the drugs are administered as single agents. For elevated liver enzymes, interrupt KEYTRUDA and axitinib, and consider administering corticosteroids as needed. With the combination of KEYTRUDA and axitinib, Grades 3 and 4 increased alanine aminotransferase (ALT) (20%) and increased aspartate aminotransferase (AST) (13%) were seen at a higher frequency compared to KEYTRUDA alone. Fifty-nine percent of the patients with increased ALT received systemic corticosteroids. In patients with ALT 3 times upper limit of normal (ULN) (Grades 2-4, n=116), ALT resolved to Grades 0-1 in 94%. Among the 92 patients who were rechallenged with either KEYTRUDA (n=3) or axitinib (n=34) administered as a single agent or with both (n=55), recurrence of ALT 3 times ULN was observed in 1 patient receiving KEYTRUDA, 16 patients receiving axitinib, and 24 patients receiving both. All patients with a recurrence of ALT 3 ULN subsequently recovered from the event.

Immune-Mediated Endocrinopathies

Adrenal Insufficiency

KEYTRUDA can cause primary or secondary adrenal insufficiency. For Grade 2 or higher, initiate symptomatic treatment, including hormone replacement as clinically indicated. Withhold KEYTRUDA depending on severity. Adrenal insufficiency occurred in 0.8% (22/2799) of patients receiving KEYTRUDA, including Grade 4 (<0.1%), Grade 3 (0.3%), and Grade 2 (0.3%) reactions. Systemic corticosteroids were required in 77% (17/22) of patients; of these, the majority remained on systemic corticosteroids. Adrenal insufficiency led to permanent discontinuation of KEYTRUDA in <0.1% (1) and withholding in 0.3% (8) of patients. All patients who were withheld reinitiated KEYTRUDA after symptom improvement.

Hypophysitis

KEYTRUDA can cause immune-mediated hypophysitis. Hypophysitis can present with acute symptoms associated with mass effect such as headache, photophobia, or visual field defects. Hypophysitis can cause hypopituitarism. Initiate hormone replacement as indicated. Withhold or permanently discontinue KEYTRUDA depending on severity. Hypophysitis occurred in 0.6% (17/2799) of patients receiving KEYTRUDA, including Grade 4 (<0.1%), Grade 3 (0.3%), and Grade 2 (0.2%) reactions. Systemic corticosteroids were required in 94% (16/17) of patients; of these, the majority remained on systemic corticosteroids. Hypophysitis led to permanent discontinuation of KEYTRUDA in 0.1% (4) and withholding in 0.3% (7) of patients. All patients who were withheld reinitiated KEYTRUDA after symptom improvement.

Thyroid Disorders

KEYTRUDA can cause immune-mediated thyroid disorders. Thyroiditis can present with or without endocrinopathy. Hypothyroidism can follow hyperthyroidism. Initiate hormone replacement for hypothyroidism or institute medical management of hyperthyroidism as clinically indicated. Withhold or permanently discontinue KEYTRUDA depending on severity. Thyroiditis occurred in 0.6% (16/2799) of patients receiving KEYTRUDA, including Grade 2 (0.3%). None discontinued, but KEYTRUDA was withheld in <0.1% (1) of patients.

Hyperthyroidism occurred in 3.4% (96/2799) of patients receiving KEYTRUDA, including Grade 3 (0.1%) and Grade 2 (0.8%). It led to permanent discontinuation of KEYTRUDA in <0.1% (2) and withholding in 0.3% (7) of patients. All patients who were withheld reinitiated KEYTRUDA after symptom improvement. Hypothyroidism occurred in 8% (237/2799) of patients receiving KEYTRUDA, including Grade 3 (0.1%) and Grade 2 (6.2%). It led to permanent discontinuation of KEYTRUDA in <0.1% (1) and withholding in 0.5% (14) of patients. All patients who were withheld reinitiated KEYTRUDA after symptom improvement. The majority of patients with hypothyroidism required long-term thyroid hormone replacement. The incidence of new or worsening hypothyroidism was higher in 1185 patients with HNSCC, occurring in 16% of patients receiving KEYTRUDA as a single agent or in combination with platinum and FU, including Grade 3 (0.3%) hypothyroidism. The incidence of new or worsening hypothyroidism was higher in 389 adult patients with cHL (17%) receiving KEYTRUDA as a single agent, including Grade 1 (6.2%) and Grade 2 (10.8%) hypothyroidism.

Type 1 Diabetes Mellitus (DM), Which Can Present With Diabetic Ketoacidosis

Monitor patients for hyperglycemia or other signs and symptoms of diabetes. Initiate treatment with insulin as clinically indicated. Withhold KEYTRUDA depending on severity. Type 1 DM occurred in 0.2% (6/2799) of patients receiving KEYTRUDA. It led to permanent discontinuation in <0.1% (1) and withholding of KEYTRUDA in <0.1% (1). All patients who were withheld reinitiated KEYTRUDA after symptom improvement.

Immune-Mediated Nephritis With Renal Dysfunction

KEYTRUDA can cause immune-mediated nephritis. Immune-mediated nephritis occurred in 0.3% (9/2799) of patients receiving KEYTRUDA, including Grade 4 (<0.1%), Grade 3 (0.1%), and Grade 2 (0.1%) reactions. Systemic corticosteroids were required in 89% (8/9) of patients. Nephritis led to permanent discontinuation of KEYTRUDA in 0.1% (3) and withholding in 0.1% (3) of patients. All patients who were withheld reinitiated KEYTRUDA after symptom improvement; of these, none had recurrence. Nephritis resolved in 56% of the 9 patients.

Immune-Mediated Dermatologic Adverse Reactions

KEYTRUDA can cause immune-mediated rash or dermatitis. Exfoliative dermatitis, including Stevens-Johnson syndrome, drug rash with eosinophilia and systemic symptoms, and toxic epidermal necrolysis, has occurred with antiPD-1/PD-L1 treatments. Topical emollients and/or topical corticosteroids may be adequate to treat mild to moderate nonexfoliative rashes. Withhold or permanently discontinue KEYTRUDA depending on severity. Immune-mediated dermatologic adverse reactions occurred in 1.4% (38/2799) of patients receiving KEYTRUDA, including Grade 3 (1%) and Grade 2 (0.1%) reactions. Systemic corticosteroids were required in 40% (15/38) of patients. These reactions led to permanent discontinuation in 0.1% (2) and withholding of KEYTRUDA in 0.6% (16) of patients. All patients who were withheld reinitiated KEYTRUDA after symptom improvement; of these, 6% had recurrence. The reactions resolved in 79% of the 38 patients.

Other Immune-Mediated Adverse Reactions

The following clinically significant immune-mediated adverse reactions occurred at an incidence of <1% (unless otherwise noted) in patients who received KEYTRUDA or were reported with the use of other antiPD-1/PD-L1 treatments. Severe or fatal cases have been reported for some of these adverse reactions. Cardiac/Vascular: Myocarditis, pericarditis, vasculitis; Nervous System: Meningitis, encephalitis, myelitis and demyelination, myasthenic syndrome/myasthenia gravis (including exacerbation), Guillain-Barr syndrome, nerve paresis, autoimmune neuropathy; Ocular: Uveitis, iritis and other ocular inflammatory toxicities can occur. Some cases can be associated with retinal detachment. Various grades of visual impairment, including blindness, can occur. If uveitis occurs in combination with other immune-mediated adverse reactions, consider a Vogt-Koyanagi-Harada-like syndrome, as this may require treatment with systemic steroids to reduce the risk of permanent vision loss; Gastrointestinal: Pancreatitis, to include increases in serum amylase and lipase levels, gastritis, duodenitis; Musculoskeletal and Connective Tissue: Myositis/polymyositis rhabdomyolysis (and associated sequelae, including renal failure), arthritis (1.5%), polymyalgia rheumatica; Endocrine: Hypoparathyroidism; Hematologic/Immune: Hemolytic anemia, aplastic anemia, hemophagocytic lymphohistiocytosis, systemic inflammatory response syndrome, histiocytic necrotizing lymphadenitis (Kikuchi lymphadenitis), sarcoidosis, immune thrombocytopenic purpura, solid organ transplant rejection.

Infusion-Related Reactions

KEYTRUDA can cause severe or life-threatening infusion-related reactions, including hypersensitivity and anaphylaxis, which have been reported in 0.2% of 2799 patients receiving KEYTRUDA. Monitor for signs and symptoms of infusion-related reactions. Interrupt or slow the rate of infusion for Grade 1 or Grade 2 reactions. For Grade 3 or Grade 4 reactions, stop infusion and permanently discontinue KEYTRUDA.

Complications of Allogeneic Hematopoietic Stem Cell Transplantation (HSCT)

Fatal and other serious complications can occur in patients who receive allogeneic HSCT before or after antiPD-1/PD-L1 treatments. Transplant-related complications include hyperacute graft-versus-host disease (GVHD), acute and chronic GVHD, hepatic veno-occlusive disease after reduced intensity conditioning, and steroid-requiring febrile syndrome (without an identified infectious cause). These complications may occur despite intervening therapy between antiPD-1/PD-L1 treatments and allogeneic HSCT. Follow patients closely for evidence of these complications and intervene promptly. Consider the benefit vs risks of using antiPD-1/PD-L1 treatments prior to or after an allogeneic HSCT.

Increased Mortality in Patients With Multiple Myeloma

In trials in patients with multiple myeloma, the addition of KEYTRUDA to a thalidomide analogue plus dexamethasone resulted in increased mortality. Treatment of these patients with an antiPD-1/PD-L1 treatment in this combination is not recommended outside of controlled trials.

Embryofetal Toxicity

Based on its mechanism of action, KEYTRUDA can cause fetal harm when administered to a pregnant woman. Advise women of this potential risk. In females of reproductive potential, verify pregnancy status prior to initiating KEYTRUDA and advise them to use effective contraception during treatment and for 4 months after the last dose.

See more here:
Merck Presents Results From Head-to-Head Phase 3 KEYNOTE-598 Trial Evaluating KEYTRUDA (pembrolizumab) in Combination With Ipilimumab Versus KEYTRUDA...

Read More...

Family of Belfast woman Eimear Gooderham (25) share memories and dealing with grief in special UTV programme – Belfast Telegraph

February 1st, 2021 4:45 pm

The story of Belfast woman Eimear Gooderham (nee Smyth), who passed away after a brave battle with cancer and sparked awareness of the stem cell register in Northern Ireland, will be told in a UTV programme this week.

imear was diagnosed with Hodgkins Lymphoma, a type of blood cancer, in 2016 aged 22 and underwent a dozen rounds of chemotherapy.

She manage to beat the cancer in the spring of 2017 and was given the all-clear by doctors, only for the disease to return again a few weeks later.

The disease went into remission following an autologous stem cell transplant, which involved using her own cells and high-dose chemotherapy.

In 2018, however, the Hodgkins Lymphoma returned once again and doctors said Eimear required another stem cell transplant, but from an anonymous donor.

This prompted her father Sean to launch a campaign, alongside UTV, to get people to sign the stem cell register and eventually a match was found.

Eimear had surgery, but sadly she passed away in hospital of organ failure on June 27, 2019, after suffering complications.

She had been due to marry her fianc Phillip Gooderham in October 2019, however with her condition worsening the wedding was organised to take place in hospital before she passed away.

UTV presenter Sarah Clarke followed Eimears story from the summer of 2018 and now that story will be told in a special programme, Eimears Wish, airing this Thursday at 10.45pm.

The programme will feature extracts from her video diary and dad Sean and sister Seainin, share memories of Eimear and talk about the positive ways they have been dealing with their grief since she passed away.

Sean Smyth said he hopes the programme will highlight the need for more people in Northern Ireland to join the stem cell donor register, especially men aged between 16 and 30.

There is also a lack of age-appropriate care for teenagers and young adults with life threatening illnesses such as blood cancer, he said.

The current facilities and the environment in which our teenagers and young adults receive their treatment and care is very poor. There also needs to be better facilities for the childrens carers.

Sarah Clarke added: It was Eimears dying wish to raise awareness of stem cell donation and to help further research into the treatment to help others. And although this programme is an entirely different one from the one we set out to make, I hope that it will in some way help to do that.

Belfast Telegraph

See the original post:
Family of Belfast woman Eimear Gooderham (25) share memories and dealing with grief in special UTV programme - Belfast Telegraph

Read More...

Disabled People Are Waiting, Anxiously, For Lifesaving Covid-19 Vaccinations – Forbes

February 1st, 2021 4:45 pm

getty

On January 26, 2021, Governor Gavin Newsom announced that California would alter its previous plan to start offering vaccines to high risk adults under 65 in the next vaccination phase. Instead, future eligibility levels in the state will be determined solely by age.

The United States Centers for Disease Controls current non-binding recommendation is to offer vaccines to disabled and chronically ill people aged 1664 years with underlying medical conditions which increase the risk of serious, life-threatening complications from COVID-19 in Phase 1c. In many states that would mean eligibility in a month or two, once the current first phases are completed. That was the plan for California, too, until this past week.

This move in California deeply disappointed the disabled community, and intensified growing concern among disabled and chronically ill people nationwide.

In a January 28 press conference, Andy Imparato, Executive Director of Disability Rights California, explained that based on current rates of vaccine production, going strictly by age will mean disabled and chronically ill people wont have access until June.

Another speaker at the virtual press conference, San Francisco disabled activist Alice Wong, has been both profoundly affected by the Covid-19 pandemic, and active in drawing attention to the unique risks and hardships the virus poses to people with disabilities and chronic illnesses. Californias change in vaccination priorities spurred her to further action:

"When I found out that Governor Newsom was eliminating prioritization for groups under Phase 1C in the state's vaccination plan I felt a surge of rage and fear at the injustice of it all. In response, I tweeted with the hashtag #HighRiskCA as a way for people from multiple communities disproportionately impacted by the pandemic to share their stories.

This is a localized variation on a hashtag thats been active since the pandemic started in March 2020, #HighRiskCovid19. Another important hashtag that has since the beginning expressed the feelings of high risk populations is #NoBodyIsDisposable.

Other disabled people also spoke out at the January 28 press conference.

Elena Escalera, Ph.D. of St. Mary's College and the #NoBodyIsDisposable Coalition said that the prospect of being included in the next phase of vaccinations is encouraging to people with disabilities and chronic illnesses. But when those priorities were changed in California to leave out people with disabilities under 65, ... there went the glimmer of hope of survival.

Anesthesiologist and bioethicist Dr. Alyssa Burgart highlighted the deep disability bias at the core of these decisions.

The bias against people with disabilities is pervasive. It is pervasive in health care because many of these folks are largely invisible. As you can see, many of these speakers have been confined to their homes because of the pandemic, and how much this has truly limited their ability to be engaged.

And Claudia Center, Legal Director of the Disability Rights Education and Defense Fund, noted the multiple layers of disability and chronic illness discrimination that disabled and chronically ill people have faced throughout the pandemic, and which also intersect with racial and other biases. These issues have included not just the latest setbacks in vaccine prioritization, but also denial of Covid-19 treatment through crisis standards of care, disabled people not being allowed to bring essential support staff with them to the hospital, and lack of data collection on how the pandemic affects disabled people.

It seems like California is making this change in priorities so it can avoid complicated and subtle decision-making, and instead go by more easily confirmed age. If so, it will achieve this simplicity by throwing some of its highest risk populations under the proverbial bus. Whatever the reasons for this change, and whether or how long its current priority system stands, it is adding to an already tense undercurrent of feeling among people with disabilities all over the country. There is a growing fear and conviction that disabled and chronically ill people, and our very specific kinds of risk from COVID-19, are once again being misunderstood and overlooked.

Obviously, everyone who isnt a vaccine or Covid skeptic is anxious to get vaccinated for the virus. And we all face the same fundamental barriers to vaccination, such as lack of sufficient supply and clumsy distribution systems. Its also important to recognize that putting any group higher on a priority list by itself doesnt do much. You can be at the top of the list, but if you cant figure out how to get a shot, or if your local provider runs out of doses, you are out of luck.

However, disabled and chronically ill people generally have more reason than most to be anxious and impatient. Some specific disabilities and conditions dont put people at that much more of a risk from Covid-19 infection, serious illness, or death, but a great many do. This is not mere speculation or paranoia. It is a documented medical fact recognized by most medical and epidemiological authorities.

Plus, being disabled exposes us to other, less direct hardships from the pandemic. For one thing, disabled people are more likely to be institutionalized in congregate care like nursing homes, assisted living, and group homes making it impossible for us to isolate ourselves. Many others of us require home care, which is less risky than nursing homes, but still exposes us to vectors of infection that we cant really do much on our own to avoid.

In a Los Angeles Times Op-Ed, Tim Jin writes:

Many people with disabilities are dealing with comorbidities of health that make us more vulnerable if we get the virus, while routine contact with multiple caregivers and other people who support us increases our risk of being exposed to COVID-19 As a person with cerebral palsy who lives on my own with support, I am more at risk because I rely on my staff to help me. I am exposed to multiple support people who come and go each day.

And Its not just people with conditions conventionally seen as disabilities who face higher risk from Covid-19. In an article for CNN, organ transplant recipient Kendall Ciesemier underscores the risk to people with chronic illnesses and other specific medical conditions:

The ones with cancer, with HIV, who have recovered from a bone marrow, stem cell, or solid organ transplant are increasingly becoming deprioritized across the country, sent to the back of the vaccine line.

She adds that these more recent setbacks only add to the sense of hopeless invisibility disabled, chronically ill, and other marginalized people have experienced throughout the pandemic:

To me and many like me, living in this pandemic has provided a daily reminder that our needs are unseen to those around us, that our lives hold little value to those who refuse to wear masks, who gather in groups or fly to a vacation destination. This is especially true for immunocompromised Black and brown people, who are among the most marginalized.

As disabled and chronically ill people we arent saying we have to be the very highest priority. We also recognize that other groups, particularly the elderly, have also at various points during the pandemic been ill-served, forgotten, or written off as acceptable losses. Most of us agree that prioritizing elderly people and healthcare workers makes sense. But we are dismayed to see disabled people who dont fit these categories seemingly forgotten.

Prioritizing everyone over 65 or 75 certainly puts some disabled people at the front of the line. But while many elderly people are also disabled, most disabled people are not elderly. According to the U.S. Census, about 34% of Americans over 65 have some kind of disability, a substantially higher disability rate than for the overall population. But only about 27% of Americans with disabilities are over 65. Disability and age overlap, but only partly.

Likewise, prioritizing health care and congregate care employees and residents is important to the disabled community, but only addresses some of us, not the vast majority who dont live in these facilities. Everyone knows about the tragedy of infection and death in nursing homes. Fewer people realize the same risk to developmentally disabled people in large institutions and smaller group homes. Meanwhile, people with disabilities who live on their own, or at home with home care, are virtually forgotten.

As a result, while we are nominally recognized to be high risk, most states vaccine priorities fail to recognize people with disabilities and chronic health conditions as a priority. Despite CDC recommendations, only 6 states currently offer vaccines to high risk adults who arent either elderly or health care / long term care workers. Many of us face the real possibility of our high-risk conditions not being recognized at all, resulting in more unnecessary illness, death, and long-term suffering.

Given the present scarcity of vaccine doses though, what is the fairer answer? This question is often presented as a false choice between deciding when to vaccinate disabled people based on science, and giving priority to the disability community for social or political reasons. In fact, it should be a combination of the two.

Scientists may know better which specific chronic illnesses and disabilities are and arent higher risk for Covid-19. But they arent always good at knowing and remembering the other ways Covid-19 disproportionately affects and endangers disabled and chronically ill people. One reason why a lot of disabled people are getting not just anxious but angry is that so many of us know from experience that without our own deliberate advocacy, its entirely possible for disabled and chronically ill people to be simply overlooked.

Deciding in a more targeted way who should have earlier access to Covid-19 vaccines is hard. Nobody is saying its not. But simply going by age, or focusing on a few specific environments and professions, isnt the answer. Its not logical, scientific, or humane.

On the other hand, the new Biden Administration appears to be a little bit ahead of the game in recognizing disabled and chronically ill peoples higher risk, and making them a higher priority. Its initial Covid-19 proposals include:

Implementation is always difficult, but a few more basic recommendations arent hard to think of. For example:

Among the many fears generated by the Covid-19 pandemic, one affecting the disabled community from the start is that our fellow Americans and portions of our government just dont care as much if we die. This idea has its roots in over a hundred years of on and off enthusiasm for eugenics the idea that society is better off without disabled people, and that disabled people themselves are, in a sense, better off dead.

A more specific fear and a profound sense of insult took hold in the early days of the pandemic when the fact that elderly and disabled people were at much higher risk of death was reported as a way to reassure other Americans that at least they werent in danger. This also turned out to be untrue, but even if it had been, it was not a proud moment in the history of American bravery or solidarity. Things only looked worse when states and localities proposed rationing policies that would deny care to people with certain kinds of disabilities who got Covid-19 explicitly and in policy writing them off.

Now its already looking like the vaccine rollout might sacrifice or simply overlook disabled people. Its probably still an exaggeration to say, as many disabled people are saying now, on social media and elsewhere, They want us dead. But the slightly less dramatic assertion that They dont care about us honestly doesnt seem far fetched these days. And even if we have our fellow Americans and governments intentions all wrong, their actions have not been promising.

Theres still time for a turnaround, but that time is running out fast.

See the article here:
Disabled People Are Waiting, Anxiously, For Lifesaving Covid-19 Vaccinations - Forbes

Read More...

Single-cell molecular profiling of all three components of the HPA axis reveals adrenal ABCB1 as a regulator of stress adaptation – Science Advances

February 1st, 2021 4:45 pm

Abstract

Chronic activation and dysregulation of the neuroendocrine stress response have severe physiological and psychological consequences, including the development of metabolic and stress-related psychiatric disorders. We provide the first unbiased, cell typespecific, molecular characterization of all three components of the hypothalamic-pituitary-adrenal axis, under baseline and chronic stress conditions. Among others, we identified a previously unreported subpopulation of Abcb1b+ cells involved in stress adaptation in the adrenal gland. We validated our findings in a mouse stress model, adrenal tissues from patients with Cushings syndrome, adrenocortical cell lines, and peripheral cortisol and genotyping data from depressed patients. This extensive dataset provides a valuable resource for researchers and clinicians interested in the organisms nervous and endocrine responses to stress and the interplay between these tissues. Our findings raise the possibility that modulating ABCB1 function may be important in the development of treatment strategies for patients suffering from metabolic and stress-related psychiatric disorders.

The hypothalamic-pituitary-adrenal (HPA) axis is pivotal for the maintenance of homeostasis in the presence of real or perceived challenges (1, 2). This process requires numerous adaptive responses involving those of the neuroendocrine and central nervous systems (3). When a situation is perceived as stressful, the paraventricular nucleus (PVN) of the hypothalamus releases corticotropin-releasing factor (CRF) to the hypophyseal portal system, connecting the hypothalamus with the anterior pituitary gland, where it stimulates the secretion of adrenocorticotropic hormone (ACTH) into the peripheral bloodstream. In turn, upon binding to the melanocortin 2 receptor, ACTH stimulates the production and secretion of glucocorticoids (GCs) from the adrenal cortex that bind to corticosteroid receptors (4). These act as transcriptional regulators providing the necessary energy resources and behavioral (emotional and cognitive) adaptations to cope with the stressful challenge and also to exert the main negative feedback at different levels of the HPA axis. While necessary for immediate response, prolonged GC exposure can increase morbidity and mortality (5, 6). Dysregulation of the neuroendocrine stress response can have severe psychological and physiological consequences, and chronic activation of the HPA axis has been linked to stress-related disorders such as anxiety disorders, major depression, posttraumatic stress disorder, and metabolic syndrome (7). As exemplified in Cushings syndrome, endogenous overproduction of GCs has detrimental effects such as impaired glucose metabolism; infectious, musculoskeletal, and cardiovascular complications; and neuropsychiatric comorbidities (8). However, despite decades of research, the molecular underpinnings of HPA dysfunction after prolonged exposure to stress are still not fully understood. Furthermore, most of the work in the field has largely focused on investigating chronic stress effects in the brain, yet much less is known about how chronic stress exposure affects the peripheral components of the HPA axis at the molecular level (9). Recent advances in the field of genomics now allow us to obtain genome-wide data on an individual cell level. Single-cell transcriptomics thereby provide powerful insight into the complexity of different tissues by enabling the identification and characterization of molecular signatures at extraordinary resolution, which can ultimately reveal previously unidentified dimensions of cell identities and their relationships with disease (10).

In this study, using single-cell RNA sequencing (scRNA-seq), we comprehensively cataloged transcriptional changes associated with chronic stress exposure in all three components of the HPA axis. We analyzed 21,723 single cells from the PVN, the pituitary, and the adrenal gland from 10 mice across two conditions (controls, n = 5; stress, n = 5). We found cell typespecific transcriptional signatures of chronic stress adaptation across the HPA axis. We identified a novel subpopulation of stress-responsive adrenocortical cells, which play an important role in the plasticity and adaptation process associated with chronic stress exposure in the adrenal cortex. We validated our findings using mouse tissues, human adrenal samples from patients with ACTH-dependent Cushings syndrome, in vitro adrenal cell models, and peripheral cortisol and genotyping data from treatment-nave, depressed patients. Our study provides the first unbiased and systematic characterization of cell typespecific signatures of the HPA axis under baseline (unstressed) and chronic stress conditions. Furthermore, our results allow a deeper understanding of HPA axis activity and its association with stress-related and metabolic disorders. Ultimately, these findings could lead to more accurate, and more reliable, molecular signatures to monitor disease progression and efficacy of treatment.

To induce chronic activation of the HPA axis, we used the chronic social defeat stress (CSDS) model, a validated, commonly used paradigm to induce long-lasting, depression- and anxiety-like endophenotypes in mice (Fig. 1A) (11). Stress exposure resulted in hallmark features of chronically stressed mice, including reduced social interaction, as demonstrated by the social avoidance test (SAT), a significant increase in basal corticosterone (CORT) levels, enhanced adrenal weight, and reduced fur quality, which is a measure associated with decreased grooming behavior (Fig. 1, B to F) (12). Body weight was not significantly different across groups after CSDS (Fig. 1G). Notably, the natural variability shown by control (unstressed) mice in the SAT did not correlate or was indicative of any of the hallmark features of chronically stressed mice (fig. S1). Five mice from each group (controls versus stressed) were selected for molecular characterization. The PVN, pituitary, and adrenal gland from these mice were used for scRNA-seq experiments (Fig. 1H).

(A) Experimental timeline of chronic social defeat stress (CSDS) paradigm for control (n = 15) and stressed (n = 15) mice. (B) CSDS reduced interaction ratios in stressed mice during the social avoidance test. Bigger dots represent the five mice from each group selected for molecular characterization (P = 0.0084, unpaired t test, two-tailed). (C and D) Twenty-one days of social defeat exposure significantly increased (a.m.) basal corticosterone (CORT) levels (P < 0.0001, unpaired t test, two-tailed) and enhanced adrenal weight (P < 0.0001, unpaired t test, two-tailed). (E) Representative adrenal glands from control and stressed mice. Scale bars, 0.5 mm. (F) CSDS significantly reduced fur quality in stressed mice [two-way analysis of variance (ANOVA), P < 0.0001]. Coat state score: (0) no wounds, well-groomed and bright coat, and clean eyes; (1) no wounds, less groomed and shiny coat OR unclean eyes; (2) small wounds, AND/OR dull and dirty coat and not clear eyes; (3) extensive wounds, OR broad piloerection, alopecia, or crusted eyes. (G) Body weight was not significantly affected by chronic stress (two-way ANOVA, P > 0.05). (H) Experimental design for scRNA-seq experiment. Individual cell suspensions were prepared from the PVN, pituitary gland (PG), and adrenal gland (AG) from selected control (n = 5) and stressed mice (n = 5). **P < 0.01, ****P < 0.0001.

To characterize inter-and intratissue heterogeneity of the HPA axis, we sequenced the transcriptome of 21,723 single cells from the PVN, pituitary, and adrenal, obtained from both unstressed (n = 5) and chronically stressed (n = 5) mice. We systematically cataloged cell identities using Scanpy, a scalable toolkit for analyzing single-cell gene expression data (13) following best practices. Graph-based clustering was performed to group cells according to their unique gene expression profiles, and dimension reduction (UMAP, Uniform Manifold Approximation and Projection) plots were used for visualization (Fig. 2) (14). In the PVN, unsupervised cluster analysis revealed a total of 18 cell clusters with distinct gene expression signatures (Fig. 2A). We determined the identity of each cluster based on the expression of established cell typespecific markers from the literature (1520). Expression of these markers across all PVN clusters can be found in fig. S2 (A to D). The 18 clusters from the PVN were further subdivided into eight major cell types as neurons, oligodendrocytes, astrocytes, microglia, endothelial, ependymal, tanycytes, and vascular cells (Fig. 2B). In the pituitary, we identified 22 unique cell clusters across 12 populations, which were grouped into somatotropes, lactotropes, corticotropes, melanotropes, gonadotropes, thyrotropes, stem cells, Pou1f1-expressing mixed cells, macrophages, endothelial cells, vascular cells, and posterior pituitary cells (Fig. 2, C and D, and fig. S3, A and B). Last, in the adrenal gland, we identified 16 unique clusters grouped into eight major groups of cells from the zona glomerulosa, zona fasciculata, a transition zone of cortical cells, medullar cells, capsular and vascular cells, macrophages, endothelial cells, and a small cluster of unknown cells (Fig. 2, E and F, and fig. S4, A and B). Expression of the top 100 genes defining the individual clusters in each of the three tissues can be found in tables S1 to S3.

(A) Dimensionality reduction Uniform Manifold Approximation and Projection (UMAP) plot depicting 6966 single cells from the PVN of the hypothalamus. Colors represent each of the 18 Louvain groups of individual cell types labeled with an abbreviation as follows: glutamatergic neurons (nGLUT1 and nGLUT2), GABAergic neurons (nGABA1 and nGABA2), mixed neurons (nMixed), vasopressin neurons (nAVP), neuropeptides (nNeuP), oligodendrocytes (Oligo1 and Oligo2), committed oligodendrocyte progenitor cells (COPs), oligodendrocyte progenitor cells (OPCs), astrocytes, endothelial, microglia, macrophages, ependymal, tanycytes, and vascular cells. (B) Distribution and percentage of eight major cell types in the PVN (purple). (C) UMAP plot depicting 9879 single cells from the pituitary. Colors represent each of the 22 Louvain groups representing individual cell types labeled with an abbreviation as follows: somatotropes (Somato1 to Somato8), lactotropes (Lacto1 and Lacto2), corticotropes (Cortico1 and Cortico2), melanotropes, gonadotropes (Gonado1 and Gonado2), thyrotropes (Thyro), endothelial, macrophages, vascular cells, stem cells, Pou1f1 mixed cells (Pou1g1 MCs), and posterior pituitary cells (PPCs). (D) Distribution and percentage of 12 major cell types in the pituitary (green). (E) UMAP plot depicting 4878 single cells from the adrenal. Colors represent each of the 16 Louvain groups representing individual cell types labeled with an abbreviation as follows: zona fasciculata (zFasc1 to zFasc5), zona glomerulosa (zGlom1 and zGlom2), transition zone of mixed fasciculata and glomerulosa cells (tZone1 and tZone2), cycling adrenocortical cells (cACCs), macrophages 1 and 2, endothelial, medullar cells, capsular and vascular cells, and unknown cells. (F) Distribution and percentage of eight major cell types in the adrenal (blue).

Next, we performed inter- and intratissue analyses to characterize cell typespecific molecular signatures of chronic stress in all three tissues of the HPA axis. First, we assessed the distribution of cell numbers for each cluster by comparing the total number of cells from the stressed group to controls (Fig. 3, A to C, and fig. S5, A to C). In the PVN, we observed a significant decrease in the number of cells from the Glut2 (32%) and neuropeptide (25%) neuronal clusters (Fig. 3A). In the pituitary, we found a significant increase in two subclusters of somatotropes (Somato6 and Somato8, 67 and 69%, respectively) (Fig. 3B). Last, in the adrenal gland, we identified the largest and most significant changes in cell distribution between the two groups. Specifically, we observed a significant increase in the number of zona fasciculata 1 cells (82%) and macrophages 2 (70%), as well as a significant decrease in the number of zona glomerulosa 1 cells (40%) (Fig. 3C).

(A to C) Distribution of cell numbers by cluster in each condition (control versus stress). Bars represent the percentage of cells from the control and stressed group per cluster (0 to 100%). All controls (gray), PVN (purple), pituitary (green), and adrenal (blue). Fishers exact test *P < 0.05, **P < 0.01, ***P < 0.001. (D) Sixty-six DEGs in 10 clusters of the PVN. Dark purple represents neurons, purple represents glial cells, and light purple represents vascular cells. (E) Six hundred ninety-two DEGs in 17 clusters of the pituitary. Dark green represents endocrine cells, green represents support cells, and light green represents stem/progenitor cells. (F) Nine hundred twenty-two DEGs in 10 clusters of the adrenal gland. Dark blue represents endocrine cells and light blue represents support cells. Size of the circle represents the number of DEGs in each cluster for all three tissues. (G) DEGs across tissues (intertissue analysis). Sixteen DEGs in common (PVN, pituitary, and adrenal), 3 DEGs (PVN and pituitary), 6 DEGs (PVN and adrenal), and 97 DEGs (pituitary and adrenal). Fourteen DEGs exclusively in the PVN (purple), 162 DEGs exclusively in the pituitary (green), and 343 DEGs exclusively in the adrenal gland (blue). Size of the circle represents the total number of DEGs in each cluster for all three tissues. (H) Expression patterns of dysregulation across DEGs per tissue. Heatmaps represent the percentage of up- and down-regulated DEGs per cluster within the PVN (purple), pituitary (green), and the adrenal (blue). Heatmap scale, 0% (gray); 50% (white); 100% (dark purple/green/blue).

Subsequently, we performed differential expression analyses to evaluate cell typespecific molecular signatures of chronic stress. We compared differentially expressed genes (DEGs) within tissues (intratissue analysis) and found that no single gene was differentially expressed (DE) across all cell types for any of the three tissues (tables S4 to S9), suggesting that cell typespecific effects of chronic stress could be masked or diluted in alternative studies using bulk RNA-seq. In contrast, when gene expression was analyzed within cell types, interesting effects emerged. In the PVN, we identified a total of 66 DEGs in 10 of the 18 cell types (Fig. 3D). In the pituitary, our analysis revealed a total of 692 DEGs in 17 of the 22 pituitary clusters (Fig. 3E). Consistent with cell distribution changes by condition, in the adrenal gland, we also observed the largest number of DEGs. Specifically, we identified 922 DEGs in 10 of the 16 adrenal clusters, ranging from 21 to 171 DE transcripts per cell type (Fig. 3F). A full list of DEGs per cell type across all three tissues can be found in tables S4 to S6.

We further compared DEGs across tissues (intertissue analysis). First, we collapsed all DEGs per tissue and identified 39 unique DEGs in the PVN, 278 in the pituitary, and 462 in the adrenal. We then looked for common genes and found 16 DEGs across all tissues (Fig. 3G). There were also 6 DEGs in common between the PVN and the adrenal, 3 DEGs between the PVN and the pituitary, and 97 DEGs between the pituitary and the adrenal glands. In addition, there were 14 genes exclusively DE in the PVN, 162 in the pituitary, and 343 in the adrenal gland (Fig. 3G and table S10). Among the genes dysregulated across the three tissues, we found several genes coding for protein members of the GC receptor (GR) chaperone complex known to play key roles in the stress response (21). Among these were HSP90 (Hsp90aa1 and Hsp90ab1), which is responsible for the direct binding of GR to the chaperone complex; HSP70 (Hspa1a and Hspa8), which encodes the first protein that recognizes and binds newly synthesized GR; and HSP40 (Dnaja1 and Dnajb1), which mediates the interaction between GR and its chaperones (22). We also observed consistent differences between the PVN and the adrenal gland for the transcription factor Nfkbia (NFB), known to interact with GCs due to their strong anti-inflammatory properties (22, 23), and Fkbp4 (encoding for the FKBP52 protein), a major regulator of GR activity (table S10) (23, 24).

Moreover, we found that most cell populations in the PVN and the pituitary showed an up-regulation of DEGs after exposure to chronic stress, except for microglial cells (PVN), macrophages, and vascular cells (pituitary), where DEGs were down-regulated (Fig. 3H). In the adrenal gland, we noticed a different pattern of regulation with several cell types, including macrophages and adrenocortical cells, showing a down-regulation of DEGs after exposure to chronic stress (Fig. 3H), suggesting a larger range of transcriptional plasticity after chronic stress at the adrenal level. Overall, these results suggest that the most profound differences due to chronic stress in the HPA axis occur in the adrenal gland, where our intra- and intertissue analyses identified the largest number of DEGs and the most significant changes at the cell population level.

The adrenal gland is a highly dynamic organ, which can quickly adapt and regenerate in response to different types of stimuli (25). For example, the adrenal significantly increases its weight in response to chronic stress, a phenomenon that has been documented in rodents, as well as human psychiatric patients (2628). In our study, we confirmed a significant increase of the adrenal weight of mice exposed to chronic social stress (Fig. 1, D and E), and single-cell transcriptomic analyses of the adrenal gland revealed a specific population of overrepresented zona fasciculata cells within the stressed group (zFasc1) (Fig. 3C). In an attempt to further investigate zFasc1 cells and identify what makes them unique, we compared their molecular profiles against all other cells in the adrenal. Because this population was so strongly driven by stress, we reasoned that the genes defining this cell type are also important responders to chronic stress. We found that the top three genes that defined the zFasc1 population were the adenosine 5-triphosphate (ATP)binding cassette subfamily B member 1B (Abcb1b) [qval: 3.27 10146; fold change (FC): 7.4], Suprabasin (Sbsn) (qval: 4.08 1084; FC: 6.6), and the 5-reductase (Srd5a2) (qval: 7.78 1082; FC: 5.8) (Fig. 4A, table S3, and fig. S5D). These genes have been previously associated with GC transport (29, 30), cell proliferation (31), and glucose metabolism (32). To validate our findings and to rule out any potential bias introduced by single-cell dissociation methods that can affect the proportions of cells in the original intact tissue, we performed mRNA in situ hybridization of Abcb1b, Sbsn, and Srd5a2 using RNAscope in adrenal glands obtained from nave or chronically stressed mice. Consistent with our single-cell results, the expression of these genes was restricted to adrenocortical cells from the zona fasciculata (Fig. 4B). Moreover, we observed a significant increase of Abcb1b and Sbsn, but not Srd5a2, mRNA expression in the zona fasciculata of stressed mice as compared to controls (Fig. 4, C to E).

(A) UMAP plot showing the expression of the top three genes that differentiate zFasc1 from other zFasc clusters: Abcb1b, Sbsn, and Srd5a2. Cyp11b1 is expressed in all zona fasciculata cells (zFasc1 to zFasc5). (B) Expression of Abcb1b, Sbsn, and Srd5a2 is restricted to adrenocortical cells from zona fasciculata. Representative adrenal glands from control and stressed mice, showing mRNA expression (brown) of Abcb1b, Sbsn, and Srd5a2 by RNAscope. Nuclei were stained with vector hematoxylin QS (purple). Scale bars, 500 m. (C to E) Chronic stress leads to a significant increase of Abcb1b (P < 0.0002) and Sbsn (P < 0.0005), but not Srd5a2 (P = 0.9715), mRNA expression in the zona fasciculata of stressed (n = 14) as compared to control (n = 14) mice. Representative images show the percentage of mRNA expression (brown) and nuclei (purple). Scale bars, 50 m. (F) CSDS leads to cellular hypertrophy in the adrenal cortex of chronically stressed mice (P < 0.0001). Bar graphs represent the average number of nuclei from the zona fasciculata. Average cell area was calculated by dividing the number of nuclei by the total area. Values are multiplied by 1000 for graphical representation. (G and H) Hypertrophy in the adrenal cortex is associated with higher levels of Abcb1b mRNA expression. Bar graphs represent the average number of nuclei present in areas of high Abcb1b (P < 0.0001) and Sbsn (P = 0.9628) mRNA expression as compared to low expressing regions in zona fasciculata of stressed mice (n = 11). All unpaired t tests, two-tailed. ***P < 0.001, ****P < 0.0001.

Subsequently, we tested whether the increase in adrenal weight after chronic stress exposure was due to an increase in the number of cells (hyperplasia) or an increase in the size of cells (hypertrophy) at the adrenal cortex. Our analysis revealed that, in stressed mice, the number of nuclei present in the zona fasciculata was significantly lower as compared to controls (Fig. 4F), suggesting cellular hypertrophy in the adrenal cortex of chronically stressed mice. Last, we evaluated whether growth characteristics of zona fasciculata cells with high expression levels of Abcb1b or Sbsn were different from low-expressing cells. Unexpectedly, our analysis revealed that the number of nuclei present in areas with high Abcb1b expression was significantly lower than in regions with low Abcb1b expression (Fig. 4G). We did not find any differences in nuclei density between regions of high or low Sbsn expression (Fig. 4H). These results suggest that hypertrophy in the adrenal cortex is associated with higher levels of Abcb1b mRNA expression.

Next, we investigated how the adrenal expression levels of Abcb1b, Sbsn, and Srd5a2 change over time, during 21 days of chronic stress exposure. Therefore, we exposed six groups of mice to a different number of social defeat sessions (0, 3, 5, 10, or 21 days). An additional group of mice received 21 days of social defeat, followed by 48 hours of recovery time to match the end point of our original CSDS paradigm (23 days) (Fig. 5A). We observed a significant and gradual increase in adrenal weight across time points (Fig. 5B). Three days of social defeat were sufficient to stimulate a significant increase in adrenal weight, which continued steadily and plateaued between days 10 and 21. We then quantified bulk mRNA expression levels of Abcb1b, Sbsn, and Srd5a2 in the adrenal cortex from these mice using quantitative real-time polymerase chain reaction (qRT-PCR). We found a significant increase of Abcb1b mRNA levels after 5 days of social defeat, while an increase for Sbsn was present only after 21 days (Fig. 5C). Consistent with our in situ nuclei quantification, we also identified a significant correlation between adrenal weight gain and the expression levels of Abcb1b (r = 0.73; P < 0.0001) and Sbsn (r = 0.51; P = 0.004) (Fig. 5D), suggesting that increases in the expression of these genes were proportional to increases in adrenal weight. In contrast, Srd5a2 did not yield any significant results in these experiments (Fig. 5, C and D). We did not find any significant differences in adrenal weight or mRNA expression levels of these genes between days 21 and 23, suggesting that the long-lasting effects of the CSDS paradigm are still present 48 hours after the last defeat session. Last, our results suggest that chronic stress exposure causes zona fasciculata cells to enlarge and increase their expression of Abcb1b, perhaps as a mechanism to cope with the increased production of GCs in the system.

(A) Experimental timeline. Six different groups of mice (n = 5) were exposed to a different number of social defeat sessions. (i) Controlno defeat, (ii) 3 days, (iii) 5 days, (iv) 10 days, (v) 21 days, and (vi) 21 days, followed by 48 hours of recovery time. (B) Three days of social defeat are sufficient to stimulate a significant increase in adrenal weight, which continued steadily and plateaued between days 10 and 21 (P < 0.0001). (C) Social defeat exposure leads to a significant increase of Abcb1b mRNA levels after 5 days of social defeat (P < 0.0001), an increase of Sbsn after 21 days (P < 0.001), while no significant changes in Srd5a2 expression (P = 0.12). Bar graphs represent mRNA levels of Abcb1b, Sbsn, and Srd5a2 normalized to Hprt. qRT-PCR, quantitative real-time polymerase chain reaction. (D) Social defeat leads to a significant correlation between adrenal weight gain and mRNA levels of Abcb1b (Pearson r = 0.73; P < 0.0001) and Sbsn (r = 0.51; P = 0.004), but no correlation with Srd5a2 expression (r = 0.29; N.S., no significance. P > 0.05). ***P < 0.001, ****P < 0.0001. CTRL, controls; SD, social defeat; D, day.

The Abcb1 gene, also known as multidrug resistance protein 1 (MDR1) or P-glycoprotein 1 (P-gp), is a well-characterized, ATP-dependent efflux pump, whose role is to transport xenobiotics and endogenous cellular metabolites across cellular membranes (33). The protein product of Abcb1 is encoded by two gene variants in mice (Abcb1a and Abcb1b) but only one gene in humans (ABCB1) (34). Moreover, it has been hypothesized that this gene modulates HPA axis activity and mediates antidepressant treatment response by regulating access of GCs and antidepressants into the brain (35). Most of the current literature in biological psychiatry has been primarily focused to understand the activity of Abcb1a in the brain, based on early observations that, in humans, the ABCB1 gene is highly expressed in endothelial cells of the blood-brain barrier (36). However, translational studies in rodents have not been successful in explaining how Abcb1 regulates the response to stress or antidepressant treatment (37). One of the reasons might be that most of these studies were carried out under the assumption that Abcb1a and Abcb1b have similar patterns of expression in the brain. Our single-cell analysis shows a very different picture with limited coexpression among the two variants. Abcb1a is specifically expressed in endothelial cells from the PVN and the pituitary (Fig. 6A), while Abcb1b is expressed in microglia and macrophages of all three tissues, in lactotropes and somatotropes of the pituitary, and in a subsection of zona fasciculata cells (zFasc1), where it shows its highest expression (Fig. 6A). Furthermore, we quantified the expression of Abcb1a and Abcb1b using publicly available bulk RNA-seq data from 35 different mouse tissues (38) and found that their expression also differs considerably in other peripheral organs. Abcb1a is lowly expressed in the periphery, while Abcb1b is the predominant variant showing high expression levels among multiple tissues, particularly in the adrenal gland where the expression of Abcb1b is several magnitudes higher as compared to every other tissue tested (Fig. 6B). These findings suggest that the adrenal gland is an important site for Abcb1 activity.

(A) UMAP plots representing cell typespecific mRNA expression of Abcb1a and Abcb1b in the PVN, pituitary, and adrenal gland of mice. (B) Bulk RNA sequencing data from 35 different mouse tissues showing mRNA expression levels of Abcb1a and Abcb1b. Heatmaps represent expression levels (0 to 12). Red, high expression; white, low expression. Expression values are displayed as Transcripts per Kilobase Million (TPM) and are log2-transformed.

Previous studies in rodents have shown that in vivo inhibition of Abcb1 by intraperitoneal injection of tariquidar, a highly specific and potent Abcb1a/b inhibitor (39), leads to a decrease in CORT levels after acute stress (40). Others have shown that mutant mice lacking both variants (Abcb1a/b) have lower baseline CORT levels as compared to wild-type controls (41). However, these studies could not attribute changes in CORT to a specific Abcb1 variant (Abcb1a or Abcb1b), nor could they conclude that the effects are modulated at the level of the brain or any of the peripheral tissues where Abcb1a and Abcb1b are expressed. To specifically explore the function of Abcb1b in the adrenal gland, we examined whether pharmacological inhibition by tariquidar modulates secretion of CORT in vitro, using an adrenocortical cell line. Mouse Y1 cells were stimulated for 24 hours with 10 nM forskolin alone, or in combination with different concentrations of tariquidar. Forskolin induces secretion of CORT by stimulation of adenylate cyclase (42). While 24 hours of forskolin treatment significantly increased CORT levels as compared to controls, we found a dose-dependent decrease of CORT with increasing concentrations of tariquidar (Fig. 7A), suggesting that GC secretion from adrenocortical cells might be dependent on Abcb1b function. In an attempt to translate our findings to humans, we assessed the modulatory role of ABCB1 on GCs, using human NCI-H295R adrenocortical cells, a validated in vitro model for steroid profiling based on their ability to produce and secrete the major steroidogenic enzymes of the adrenal cortex (43). In line with our previous results, treatment of NCI-H295R cells with 10 nM forskolin led to a significant increase of medium cortisol levels, as compared to vehicle-treated controls (Fig. 7B). Treatment with increasing concentrations of tariquidar led to a significant decrease of media cortisol levels (Fig. 7B). Together, our results show that in vitro pharmacological manipulation of Abcb1 in adrenocortical cell lines leads to a decrease in GC secretion, suggesting that modulation of Abcb1b in adrenocortical cells affects GC secretion in both mice and humans.

(A) Pharmacological inhibition of Abcb1 by tariquidar in mouse Y1 adrenocortical cells. CORT levels (ng/ml) after 24-hour treatment with forskolin (0 and 10 nM) or tariquidar (0, 10, 50, 125, 250, 500, and 1000 nM). (B) Pharmacological inhibition of ABCB1 by tariquidar in human NCI-H295R adrenocortical cells. Cortisol levels (ng/ml) after 24-hour treatment with forskolin (0 and 10 nM) or tariquidar (0, 10, 50, 125, 250, 500, and 1000 nM). One-way ANOVA, ****P < 0.0001.

In humans, chronic endogenous oversecretion of ACTH due to a pituitary or ectopic tumor results in excessive GC secretion and enlargement of the adrenal glands (Fig. 8A) (44). Thereby, this disease stage overlaps with the chronic activation of the HPA axis and hypersecretion of GCs in stress-related disorders. In cases of unsuccessful pituitary surgery or in those patients in whom the ectopic source of ACTH remains obscure, bilateral adrenalectomy is required to treat steroid excess. This opened the possibility to study adrenal glands that had been chronically stimulated and to compare those with controls in the absence of ACTH oversecretion. We quantified mRNA expression of ABCB1 and SBSN, using RNAscope in cases (n = 8) and controls (n = 6). SBSN mRNA was not detectable in human adrenocortical samples (Fig. 8B). Following this approach, we identified a significant up-regulation of ABCB1 mRNA in Cushings adrenocortical samples, as compared to controls (Fig. 8C). These results are consistent with our initial findings in chronically stressed mice and reinforce our evidence for a role of ABCB1 as a modulator of GC activity in the adrenal gland. In addition, our results highlight ABCB1 as a potential regulator of the detrimental effects of impaired glucose metabolism associated with patients with Cushings syndrome.

(A) Graphical representation of the effects of Cushings disease on the adrenal gland. (B and C) Expression of ABCB1 (P = 0.0056) and SBSN (P = 1.0) mRNA, using RNAscope in adrenal glands from patients with ACTH-dependent Cushings syndrome (n = 8) and controls (n = 6). Representative images show the percentage of mRNA expression (brown) and nuclei (purple) normalized by total area. Scale bars, 50 m, **P < 0.01.

Individuals who lack ABCB1, as it occurs in some breeds of dogs with the ABCB1-1 mutation (45), have severe adverse reactions to common medications that act as substrates of this transporter, such as immunosuppressants and steroid hormones (46). Previous studies have shown that dogs and rodents lacking a functional Abcb1/ABCB1 gene have a blunted HPA axis response compared to wild-type animals (41, 47). In humans, multiple single-nucleotide polymorphisms (SNPs) map to the ABCB1 gene locus, and some of these variants have been associated with reduced protein function and activity (48). One of the most studied ABCB1 polymorphisms is the rs2032582 (G2677T), which is a nonsynonymous variant on exon 21 that has been linked to major depressive disorder and treatment response (49). To investigate the relevance of our findings in depressed human patients, we examined whether the ABCB1 polymorphism rs2032582 is associated with an altered HPA axis response, using peripheral plasma samples from 154 treatment-nave, depressed patients. We measured plasma ACTH and cortisol concentrations in depressed patients at baseline, following CRF stimulation, and 15-min intervals for the following hour (Fig. 9A). The genotype and allele distributions of rs2032582 in patients are shown in Fig. 9A. At baseline, patients with the minor allele (TT) showed a decrease in cortisol levels as compared to the major (GG) and heterozygote (TG) alleles; however, this effect did not reach statistical significance after Bonferroni correction (Fig. 9B). After CRF stimulation, we found a significant genotype-by-time interaction in patients cortisol levels (qval: 0.033). More specifically, patients with the minor (TT) allele showed a dampened cortisol response after CRF stimulation (Fig. 9C). We did not find any statistical differences in ACTH levels after CRF stimulation (Fig. 9D), suggesting that the effects of rs2032582 on the ABCB1 gene might be taking place at the level of the adrenal gland. These results are consistent with our mouse and cell culture findings and support the idea that Abcb1/ABCB1 function may regulate HPA axis response.

(A) Experimental design. Predictors of remission in depression to individual and combined treatments (PReDICT) cohort (N = 154) to investigate effects of the ABCB1 variant rs2032582 on HPA axis function. CRF stim, CRF stimulation test; SNP, single-nucleotide polymorphism; HWE-P, Hardy-Weinberg equilibrium P value. (B) Baseline cortisol levels (g/ml) for treatment-nave, depressed patients carrying the rs2032582 SNP genotype. (C and D) Cortisol (g/ml) and ACTH (pg/ml) levels after CRF stimulation (log-transformed). Only completers were included in the analysis. There are no dropouts in sample sizes over time for cortisol or ACTH. GG = major homozygotes (n = 56), GT = heterozygotes (n = 74), TT = minor homozygotes (n = 24). Mixed effects models, Bonferroni-corrected *P < 0.05.

Despite decades of research, the molecular and cellular identity of the HPA axis components, their inter-relationships, and their function after chronic stress exposure are still only partially understood. Here, using scRNA-seq, we describe cell typespecific molecular signatures of chronic stress in all three components of the HPA axis, providing a level of resolution never before reached.

The PVN integrates and coordinates the neuroendocrine HPA axis response to stressful stimuli. However, aside from containing the neuroendocrine neurons that control the synthesis and release of CRF, the PVN also exhibits a significant degree of cellular and molecular complexity, with multiple types of neuronal and nonneuronal subtypes. In this study, we characterized and described the cellular heterogeneity and identity of all cell types in the PVN. We identified many DEGs that are involved in the intracellular trafficking of the GC and mineralocorticoid receptors and play key roles in the response to chronic stress, such as heat shock proteins and Fkbp4 across multiple cell types (24). We also found groups of genes that were DE in unique cell types, such as the cysteine-rich angiogenic inducer 61 gene (Cyr61), which was only found DE in ependymal cells. Cyr61 is a target gene of the hippo signaling pathway, which regulates tissue homeostasis, regeneration, proliferation, and growth and has recently been linked to the pathophysiology of stress-related psychiatric disorders (50). In the neuropeptide cluster, we found a down-regulation of corticotropin-releasing factor (Crf) and vasopressin (Avp), as well as an up-regulation of oxytocin (Oxt) and somatostatin (Sst); however, these changes did not survive correction for multiple testing. We did not find any significant dysregulation of GR (Nr3c1) mRNA in any of the clusters of the PVN. Nevertheless, we did find a significant difference in the total number of Nr3c1+ cells in some of the cell clusters of the PVN (fig. S2D), suggesting that the GR mRNA differences reported in the literature (4, 6, 9) could be due to a decrease in the total number of Nr3c1+ cells after chronic stress, rather than lower expression levels of the existing cells. A decrease in the total number of Nr3c1+ cells is not found across all cell types of the PVN but is rather limited to specific cell populations. These populations could represent the cell types where stress exerts its main effects in the PVN via GR. However, these findings would need to be further validated and replicated in other studies. Last, we found that most cell populations across the PVN showed an up-regulation of DEGs after exposure to chronic stress, except for microglial cells where most DEGs were down-regulated. These changes in microglial cells in combination with gene expression differences (across multiple cell types) of several genes involved in the intracellular trafficking of GCs are possibly the result of overexposure to GCs during a prolonged (chronic) stress paradigm. GCs are released during the stress response and are well known for their immunosuppressive and anti-inflammatory properties. In addition, growing evidence suggests that changes in neuroendocrine function and metabolism are significant triggers of inflammation, which has been linked to the development of neuropsychiatric disorders. Ultimately, while this is an important issue in the field, it is logistically challenging to address considering that the effects or stress, GCs, and inflammation are closely intertwined, likely powering each other in a bidirectional way.

The second component of the HPA axis, the pituitary gland, is a complex organ and an important regulator of major physiological processes, including the neuroendocrine stress response (51). It is composed of a heterogeneous mix of endocrine, general support, and stem cells (17, 18). Despite a significant body of research characterizing attributes of pituitary activity, the cell typespecific regulation of chronic stress at the pituitary level is still poorly understood. Here, we characterized cell typespecific molecular signatures of chronic stress in the pituitary gland. Among many, our DE analysis revealed several genes that were consistently dysregulated in multiple endocrine cells, such as somatotropes, gonadotropes, lactotropes, and corticotropes. Specifically, we found an up-regulation of Cd63, Hsp90aa1, and Hsp90ab1, as well as a down-regulation of several ribosomal genes in all four cell types, suggesting altered GC and ribosomal activity. Moreover, corticotropes are directly stimulated by CRF and are responsible for the release of ACTH into circulation. In our analysis, we found 32 DEGs in this population. However, we did not find any significant differences in the expression of the corticotropin-releasing hormone receptor 1 (Crhr1) or the GR (Nr3c1) gene. Furthermore, and consistent with our findings in the PVN, we found that most cell types across the pituitary showed an up-regulation of DEGs after exposure to chronic stress, except for macrophages and vascular cells, where most DEGs are down-regulated. In our single-cell data, we found a large number of DEGs across multiple types of endocrine cells, suggesting that the stress response in the pituitary gland is a dynamic and complex process that is not limited to the effect that CRF exerts on corticotropes. In our analyses, somatotropes were the population of pituitary cells that showed the biggest changes after chronic stress, both in terms of number of DEGs and changes in proportions of cells. Somatotropes produce and release growth hormone, and they play an important role in the regulation of GC synthesis and adrenal growth and have been shown to positively affect adrenal cell size and number of adrenocortical cells (52). However, the role that somatotropes play in chronic stress and the development of stress-related psychiatric disorders are still poorly understood. Our high-throughput, cell typespecific findings of the effects of chronic stress on the pituitary and somatotropes are both novel and a significant advancement to our understanding of the mechanisms of stress adaptation in the pituitary gland.

Last, the adrenal gland is a major effector of the HPA axis, where interplay between several types of specialized cells takes place to coordinate a complex endocrine, immune, and metabolic response to stress. It is composed of the adrenal medulla and the adrenal cortex, two embryonically different endocrine tissues (25). The adrenal cortex is further divided into three major zones: zona glomerulosa (zG), zona fasciculata (zF), and zona reticularis (zR), each responsible for the synthesis and release of mineralocorticoids, GCs, and androgens, respectively (27). Zona reticularis has been shown to be absent in mice (25). Until now, our understanding of the mechanisms responsible for chronic stress adaptation in the adrenal has been limited. Our study is the first to provide a cell typespecific, unbiased, molecular characterization of the adult adrenal gland (under baseline or chronic stress conditions). Across several cell types, we found a significant dysregulation of genes coding for steroidogenic enzymes responsible for the biosynthesis of corticosteroids, such as GCs and mineralocorticoids. More specifically, we found a dysregulation of Star, Fdx1, Cyp11b1, Cyp21a1, Cyp11a1, Hsd3b1, Nr4a1, and Agtr1a after exposure to chronic stress. In contrast to what we found in the PVN and pituitary, cell types in the adrenal showed both up-regulation and down-regulation of DEGs after exposure to chronic stress, suggesting a larger range of transcriptional plasticity after chronic stress at the adrenal level. Although the changes in the expression of genes coding for steroidogenic enzymes are consistent with the current literature (27), our results offer a new level of resolution by describing the specific cell types where these changes take place in the adrenal. Furthermore, our results highlight that changes after chronic stress in the adrenal are not limited to the endocrine cells of the adrenal cortex or adrenal medulla. In our data, we also find significant changes in the number of macrophages, as well as the number of DEGs in this cluster, after chronic stress. Macrophages are modulated by GCs to secrete cytokines and regulate inflammation and the immune system (53). To the best of our knowledge, this study is the first to show a significant effect of chronic stress in macrophages of the adrenal gland. In addition, our results show a global dysregulation of transcriptional activity in macrophages across all three components of the HPA axis (PVN, pituitary gland, and adrenal gland), suggesting that this cell population is part of a common, multilevel and multitissue signaling network that regulates adaptation to chronic stress.

One of the main findings from our study is the identification of a novel population of overrepresented Abcb1b+ cells within the zona fasciculata of the stressed group. The identification of this novel and specialized cell type in the adrenal gland could not have been possible using standard bulk RNA-seq methods. All previous transcriptomic studies examining the effects of chronic stress in the adrenal gland have been limited to adrenocortical, adreno-medullar, or whole tissue homogenates that average out the signature of thousands of cells, which can mask, dilute, or even distort signals of interest coming from specialized cell populations. Hence, one can expect that any cell typespecific signature of chronic stress (as is the case for zFasc1 cells) has been diluted or even lost in these studies. Here, through a series of complementary experiments, we validated this novel subpopulation of Abcb1b+ cells in the adrenal cortex, which play an important role in stress adaptation. Our experiments showed that increased mRNA expression of Abcb1b+ cells in the adrenal gland is associated with increased adrenal weight and cellular hypertrophy in the adrenal cortex of stressed mice, suggesting that chronic stress exposure causes zona fasciculata cells to enlarge and increase their expression of Abcb1b, perhaps as a mechanism to cope with the increased and sustained production of GCs in the system. The Abcb1 gene is a well-characterized efflux pump whose role is to transport substances, deemed as harmful, across membranes. However, most of the work to study this gene in psychiatry has been primarily focused on understanding the activity of the variant Abcb1a in the brain. Our single-cell analysis in combination with bulk RNA-seq data from 35 different mouse tissues showed that Abcb1b is the predominant variant in the periphery showing high expression levels among multiple tissues, particularly in the adrenal gland, suggesting that the adrenal is an important site for Abcb1 activity. Furthermore, to disentangle the effects of Abcb1a versus Abcb1b in the response to stress, we performed a series of in vitro experiments in mouse and human adrenocortical cells. Our results showed that pharmacological inhibition of Abcb1b in adrenocortical cell lines leads to a decrease in GC secretion, suggesting that modulation of Abcb1b in adrenocortical cells affects GC activity in both mice and humans. Moreover, in an attempt to translate our findings to humans, we investigated the expression of ABCB1 in adrenal cortical tissues from patients diagnosed with ACTH-dependent Cushings syndrome. These patients suffer from excessive GC secretion and adrenal hypertrophy due to a pituitary or ectopic tumor. Thus, this disease stage overlaps with the chronic activation of the HPA axis and hypersecretion of GCs in stress-related disorders. We found a significant up-regulation of ABCB1 in cases, as compared to controls. In addition to being consistent with our findings in chronically stressed mice, these results highlight the role of ABCB1 as a modulator of GC activity in the adrenal gland and postulate ABCB1 as a potential regulator of the impaired glucose metabolism associated with Cushings syndrome. Last, we investigated the relevance of our findings in depressed human patients by examining whether the ABCB1 polymorphism rs2032582 (G2677T) is associated with an altered HPA axis response in peripheral plasma samples from treatment-nave, depressed patients. In humans, the rs2032582 polymorphism has been associated with reduced protein function and activity and has been linked to major depressive disorder, suicidal ideation, and treatment response (49). Consistent with our findings in mice, adrenocortical cell lines, and adrenocortical samples from human Cushings patients, we found that, after CRF stimulation, patients with the minor (TT) allele showed a dampened cortisol but normal ACTH response, suggesting that the effects of rs2032582 on the ABCB1 gene might be taking place at the level of the adrenal gland. In addition, our results support the idea that Abcb1/ABCB1 function may regulate HPA axis response.

Together, our data offer new insights into how chronic stress regulates transcriptional activity in a multilevel, cell typespecific fashion. We identified hundreds of novel genes that are dysregulated across all tissues and levels of the HPA axis. On the basis of our intra- and intertissue analyses, we found the most profound differences due to chronic stress in the adrenal gland, which had the highest number of DEGs and the most significant changes at the cell population level. Through a series of complementary behavioral, molecular, cellular, and functional experiments, we identified a novel subpopulation of Abcb1b+ cells in the adrenal cortex, which play an important role in the adaptation process and plasticity associated with chronic stress exposure. The exact mechanism underlying the effect of ABCB1 on GC regulation and secretion in the adrenal cortex still needs to be further explored. However, previous studies have shown that transcriptional regulation of the Abcb1 genes can be mediated through a putative GC response element (GRE) identified in the promoter region of these genes in both rodents and humans (54, 55). At least in mice, this GRE binding site is only found in the promoter region of the Abcb1b variant, suggesting that Abcb1b is directly regulated by GCs in the periphery, predominantly in the adrenal glands. Therefore, we propose that the Abcb1b/ABCB1 gene and protein are involved in mediating chronic stress adaptation through regulation and control of GCs in the adrenal gland. Our findings raise the prospect that modulating ABCB1 function may be important in the treatment of patients suffering from neuropsychiatric and metabolic disorders, such as stress-related disorders and Cushings syndrome. They further suggest that adrenal ABCB1 activity could be used to stratify patients and tailor treatment strategies. Ultimately, our results provide a deeper understanding of the complex mechanisms of HPA axis regulation.

All experiments were performed in accordance with the European Communities Council Directive 2010/63/EU. All protocols were approved by the Ethics Committee for the Care and Use of Laboratory Animals of the government of Upper Bavaria, Germany. Male mice aged between 7 and 10 weeks old were used for all experiments. Mice were bred in the animal facility of the Max Planck Institute of Biochemistry (Martinsried, Germany) and group-housed (four to five mice per cage) until 1 week before the start of the experiments, when mice were single-housed. Mice were kept in individually ventilated cages (IVCs; 30 cm by 16 cm by 16 cm; 501 cm2), serviced by a central airflow system (Tecniplast, IVC Green LineGM500), according to institutional guidelines. IVCs had sufficient bedding and nesting material as well as a wooden tunnel for environmental enrichment. Animals were maintained under pathogen-free, temperature-controlled (23 1C), and constant humidity (55 10%) conditions on a 12-hour light/12-hour dark cycle (lights on at 7:00 a.m.) with food and water provided ad libitum, at the Max Planck Institute of Psychiatry (Munich, Germany).

C57BL/6N males (7 weeks old) were exposed to the CSDS paradigm for 21 consecutive days, as previously described (12). Briefly, experimental mice were introduced daily into the home cage of a dominant CD1 resident mouse, which rapidly recognized and attacked the intruders within 2 min. To avoid serious injuries, the subordinate mouse was separated immediately after being attacked by the CD1 aggressor. After the physical encounter, mice were separated by a perforated metal partition, allowing the mice to keep continuous sensory but not physical contact for the next 24 hours. Every day, for a total of 21 days, mice were defeated by another unfamiliar, CD1 resident mouse, to exclude a repeated encounter throughout the experiment. Defeat encounters were randomized, with variations in starting time (between 9:00 a.m. and 6:00 p.m.) to decrease the predictability to the stressor and minimize habituation effects. Control mice were single-housed, in the same room as the stressed mice, throughout the course of the experiment. All animals were handled daily and weighed every 4 days. Coat state was scored on a scale of 0 to 3 according to the following criteria: (0) No wounds, well-groomed and bright coat, and clean eyes; (1) no wounds, less groomed and shiny coat, OR unclean eyes; (2) small wounds, AND/OR dull and dirty coat, and not clear eyes; (3) extensive wounds, OR broad piloerection, alopecia, or crusted eyes. End point and tissue collection were performed in the morning (9:00 a.m.) and 48 hours after the last social defeat session (day 23). This was done to capture the cumulative effects of chronic stress, rather than the acute effects of the last defeat session. The SAT was conducted during the last week of the CSDS paradigm, and based on their performance, five mice from each group were selected for molecular characterization, thus avoiding potentially stress-resilient animals. For end point, all mice were deeply anesthetized with isoflurane and perfused with cold phosphate-buffered saline (PBS), and target tissues were quickly dissected for molecular experiments. Cardiac blood was collected for the assessment of basal CORT levels. Adrenal glands were dissected from fat and weighed. The brains, pituitary, and adrenal glands from selected mice were immediately processed for RNA single-cell analysis. Tissues from all remaining mice were collected for downstream validation experiments.

Social avoidance behavior was assessed with a novel CD1 mouse in a two-stage social interaction test. In the first 2.5-min test (nontarget), the experimental mouse was allowed to freely explore the open-field arena containing an empty wire mash cage against one wall of the arena (labeled as the interaction zone). In the second 2.5-min test (target), the experimental mouse was returned to the arena with an unfamiliar male CD1 mouse enclosed in the wire mash cage. The ratio between the time in the interaction zone of the nontarget trial and the time in the interaction zone of the target trial was calculated and deemed as the interaction time ratio.

Blood sampling was performed during end point (9:00 a.m.) by collecting blood from the heart of each mouse before perfusion with PBS. All blood samples were kept on ice and centrifuged at 4C, and 10 l of plasma was removed for measurement of CORT. All plasma samples were stored at 20C until CORT measurement. CORT concentrations were quantified by radioimmunoassay (RIA) using a CORT double antibody 125I RIA kit (sensitivity: 25 ng/ml; MP Biomedicals Inc.) following the manufacturers instructions. Radioactivity of the pellet was measured with a gamma counter (Wizard2 2470 Automatic Gamma Counter; Perkin Elmer). All samples were measured in duplicate, and the intra- and interassay coefficients of variation were both below 10%. Final CORT levels were derived from the standard curve.

Six different groups (each N = 5) of C57BL/6N males were exposed to a different number of social defeat sessions to assess the cumulative effects of stress and its correlation with changes in mRNA levels of Abcb1b, Sbsn, and Srd5a2. The groups were defined as follows: (i) controlno defeat, (ii) 3 days of social defeat, (iii) 5 days of social defeat, (iv) 10 days of social defeat, (v) 21 days of social defeat, and (vi) 21 days of social defeat, followed by 48 hours of recovery time. The last group was introduced to match the end point of our original chronic social defeat paradigm cohort (23 days). All mice were 7 weeks old at the beginning of the experiment. Individual social defeat encounters were carried out exactly as previously explained in the CSDS paradigm section of the methods. End point and tissue collection were performed in the morning (9:00 a.m.) of day 23. All mice were deeply anesthetized with isoflurane, and the adrenal glands were quickly dissected for molecular experiments. Adrenal glands were further dissected from fat and weighed. Trunk blood was collected for the assessment of basal CORT levels.

Tissue dissociation. Mice were anesthetized lethally using isoflurane and perfused with cold PBS to get rid of undesired blood cells in target tissues. Brains, pituitaries, and adrenal glands were quickly dissected and immediately transferred to ice-cold oxygenated artificial cerebral spinal fluid (aCSF) (brains), ice-cold Hanks balanced salt solution (HBSS) (pituitaries), or ice-cold PBS (adrenals) and kept in the same solutions during dissection and dissociation. The aCSF was oxygenated throughout the experiment with a mixture of 5% CO2 in O2. Sectioning of the brain was performed using a 0.5-mm stainless steel adult mouse brain matrix (Kent Scientific) and a Personna Double Edge Prep Razor Blade. A slide (approximately 0.58 mm Bregma to 1.22 mm Bregma) was obtained from each brain, and the extended PVN was manually dissected under the microscope. Two cell suspensions were prepared for each of the three tissues with one pool for controls and one pool for stressed mice. The PVN from five different mice was pooled and dissociated for 35 min using the Papain Dissociation System (Worthington) following the manufacturers instructions. Similarly, the pituitaries from five mice were pooled and dissociated for 15 min using papain. Last, the adrenal glands from five mice were pooled and dissociated for 55 min using a 0.2% collagenase II solution. All cell suspensions were incubated at 37C using a shaking water bath. After this, cell suspensions were filtered with 30-m filters (Partec) and kept in cold aCSF, HBSS, or PBS.

Cell capture, library preparation, and high-throughput sequencing. Cell suspensions with approximately 1,000,000 cells/l were used. Each pool was loaded onto individual lanes of a 10X Genomics Chromium chip, as per factory recommendations. For all three tissues, the control and stress cell suspensions were loaded and processed together in the same chip to avoid batch effects by condition. Reverse transcription and library preparation were performed using the 10X Genomics Single-Cell v2.0 kit following the 10X Genomics protocol. Molar concentration and fragment length of libraries were quantified by qPCR using KAPA Library Quant (Kapa Biosystems) and Bioanalyzer (Agilent High Sensitivity DNA kit), respectively. Each library was sequenced on a single lane of an Illumina HiSeq4000 System generating 100base pair paired-end reads at a depth of ~340 million reads per sample.

Preprocessing and quality control. Preprocessing of the data was done using the 10X Genomics Cell Ranger software version 2.1.1 in default mode. The 10X Genomics supplied reference data for the mm10 assembly and corresponding gene annotation was used for alignment and quantification. All further analyses were performed using Scanpy (version 1.4.4.post1) (13), following guidelines from an established best practices workflow (14). For quality control, we looked at the distribution of count depth, number of genes, and mitochondrial read fraction per sample. Because distributions were fairly homogeneous, we chose to pick the same thresholds for all samples (tissues and conditions). Specifically, we filtered out (i) cells with less than 1000 counts, (ii) less than 400 genes detected, and (iii) percentage of mitochondrial gene counts higher than 25%. In addition, genes expressed in less than 20 cells were removed as well. Quality control (QC) plots can be found in fig. S5 (E to G). This resulted in a dataset of 21,723 single cells, of which 6966 cells and 16,168 genes were from the PVN, 9879 cells and 15,437 genes were from the pituitary, and 4878 cells and 13,997 genes were from the adrenal gland. The size factors used for normalization were obtained using Scran (version 1.14.5) (56), and the data were log1 Ptransformed. Each dataset was batch-corrected using Combat (57), available in Scanpy. For each tissue, we selected the top 4000 highly variable genes using the highly_variable_genes function. Dimensionality reduction was performed using principal components analysis computed on highly variable genes and taking the first 50 PCs. Last, we computed a k-nearest neighbor graph (KNN)graph (k = 15) on the low-dimensional embedding, necessary for UMAP visualization.

Clustering, marker gene identification, and cluster annotation. Data were clustered using the Louvain (version 0.6.1) algorithm implemented in Scanpy (13). This is a graph-based clustering method that relies on the KNN-graph discussed above. We clustered at two different resolution levels (r = 0.5 and r = 1). After inspection of the cell clusters, we observed that those obtained using a finer resolution (r = 1) aligned better with our annotations and therefore used them for visualization and downstream analyses. Marker genes for each cluster were detected using a Welchs t test between cells in the cluster and all cells outside of it as reference. This was done using the rank_genes_groups function implemented in Scanpy and computed on log-normalized nonbatch-corrected data. Cell types were determined using a combination of marker genes identified from the literature and Gene Ontology for cell types using the web-based tool: mousebrain.org (58).

Differential expression analysis. Differential expression analyses were performed using MAST (59) implemented in R, which models scRNA-seq data using a generalized linear model (GLM). The computation was performed on log-normalized nonbatch-corrected data, and, for each cell cluster, we fit the following model: [Y ~ 1 + condition + n_genes], where Y is the log-normalized nonbatch-corrected data, 1 is the intercept term, condition is the covariate that accounts whether the mouse was stressed or not, and n_genes is used as a technical covariate as a proxy for technical and biological factor that might influence gene expression. The test produced a P value for each gene in each cell cluster and a q value, which is the P value after adjustment for multiple testing, using false discovery rate (FDR) correction. Furthermore, the mean expression of each gene for the two different conditions was computed.

Ambient RNA assessment. After QC analyses, we noticed the presence of highly expressed genes across all cells, despite being known marker genes of specific cell types. We noticed that most of these genes coded for neuropeptides or hormones and decided to assess whether we could explain this as ambient RNA contamination. To investigate which genes were expressed as ambient RNA, we analyzed the unfiltered datasets for the three tissues. We once again looked at the count depth distribution for what we conclude are empty droplets and selected cells with counts between 100 and 300 for the PVN, between 300 and 600 for the pituitary, and between 50 and 200 for the adrenal gland. We also removed genes that are expressed in less than 20 cells. After these steps, we obtained a dataset of 120,320 droplets and 14,129 genes for the PVN, 113,043 droplets and 13,777 genes for the pituitary, and 107,698 droplets and 11,684 genes for the adrenal gland. Because these are empty droplets and we do not expect any meaningful clustering of the data, we used a less sophisticated normalization technique, normalizing each cell by total counts over all genes, thus obtaining the same total count per cell after this step. The number of counts per cell to obtain was selected automatically as the median count per cell before normalization. For all tissues, we computed the mean expression of each gene across all droplets by condition (stress versus control). Furthermore, to exclude from our list of significantly DEGs those that are detected owing to differential ambient RNA expression, we performed differential expression testing using MAST across all droplets using the same GLM formulation defined above (note that, in our previous analysis, we tested within each cluster).

For paraffin embedding, adrenal glands were dissected and the surrounding fat was removed and fixed in 10% neutral buffered formalin (Sigma-Aldrich, HT501128) overnight at room temperature. Tissue was embedded manually over 3 days. All washes were carried out for 1 hour at room temperature unless indicated. Day 1: three times PBS, 25% EtOH, 50% EtOH, 70% EtOH, and 70% EtOH overnight at 4C. Day 2: 80% EtOH, 90% EtOH, 95% EtOH, and 100% EtOH overnight at 4C. Day 3: 100% EtOH, Neoclear (Sigma-Aldrich, 109843) I for 10 min at room temperature; Neoclear II for 10 min at 60C; Neoclear: paraffin 1:1 for 15 min at 60C, paraffin I for 1 hour at 60C, paraffin II for 1 hour at 60C, and paraffin III for 1 hour at 60C. Samples were sectioned at 5 m. RNAscope was carried out on paraffin-embedded sections with the RNAscope 2.5 HD Kit-BROWN (ACD bio 322300) assay following the manufacturers protocols, with Standard timings for retrieval and protease treatment. The following probes were used (all ACD bio): Mm-Abcb1b (422191), Mm-Sbsn (564441), Mm-Srd5a2 (431361), Hs-ABCB1 (401191), and Hs-SBSN (447411). Positive control Mm-Ppib (313911), Hs-UBC (310041), and negative control dapB (310043) were also used. Nuclei were stained with Vector Hematoxylin QS (Vector Laboratories, H-3404), and slides were mounted in VectaMount Permanent Mounting Medium (Vector Laboratories, H-5000).

Paraffin sections were deparaffinized and rehydrated as per immunohistochemistry. Slides were incubated for 30 s with Hematoxylin QS, washed with running water, incubated for 30 s with eosin, washed with running water, and mounted in VectaMount Permanent Mounting Medium.

Hematoxylin and eosin and RNAscope slides were scanned with a NanoZoomer-XR digital slide scanner (Hamamatsu). Images were processed with NanoZoomer digital pathology view (Hamamatsu), and quantification was done with Fiji.

Four areas of the same dimensions (252 252 pixels) were selected from the zona fasciculata of the cortex. Nuclei were counted, and the average cell area was calculated by dividing the number of nuclei by the total area. Values were multiplied by 1000 for graphical representation.

Quantification of messenger RNA levels of Abcb1b, Sbsn, and Srd5a2 in the adrenal glands was carried out using qRT-PCR. Total RNA was reverse-transcribed using the High-Capacity cDNA Reverse Transcription Kit (Applied Biosystems). RT-PCR reactions were run in triplicate using the ABI QuantStudio 6 Flex RT-PCR System and data were collected using the QuantStudio RT-PCR software (Applied Biosystems). Expression levels were calculated using the standard curve, absolute quantification method. The endogenous expressed gene Hprt was used to normalize the data. The following Taqman probes were used: Abcb1b: Mm00440736_m1, Sbsn: Mm00552057_m1, Srd5a2: Mm00446421_m1, and Hprt: Mm00446968_m1.

Mouse Y1 cells and human NCI H295R adrenocortical cells were seeded into 12-well plates and incubated overnight using Dulbeccos modified Eagles medium high glucose (4.5 g/liter) (Gibco) with 7.5% horse serum (Gibco), 2.5% fetal bovine serum (FBS) (Gibco), and 1% penicillin-streptomycin (Gibco) and RPMI 16/40 + GlutaMax (Gibco) with 10% FBS (Gibco), 1% Insulin-Transferrin-Selenium-Ethanolamine (ITS) (Thermo Fisher Scientific), and 1% penicillin-streptomycin (Gibco), respectively. In this experiment, 100,000 Y1 and NCI H295R adrenocortical cells per well were used. Cells were then stimulated for 24 hours with 10 nM forskolin and subsequently treated with different concentrations of tariquidar (0, 10, 50, 125, 250, 500, and 1000 nM) and incubated for 24 hours. Last, supernatants and cell pellets were collected and harvested for further analyses and measurement of CORT (ng/ml) and cortisol (g/liter) levels. Media CORT levels in Y1 cells were quantified by RIA using a CORT double antibody 125I RIA kit, as previously described in the animal experiments. Media cortisol levels in NCI H295R adrenocortical cells were determined using an enzyme-linked immunosorbent assay (ELISA) kit (RE52061, TECAN, IBL Hamburg, Germany). The standard range was 20 to 800 ng/ml.

The study was approved by the Ethics Committee of the University of Wuerzburg (Germany) (#88/11), and written informed consent was obtained from all subjects. Eight patients with biochemically confirmed persistent ACTH-dependent Cushings syndrome were studied. Cushings syndrome was established according to current guidelines (60). Half of the patients (n = 4) had pituitary-dependent Cushings syndrome, while, in the other patients (n = 4), ectopic Cushings syndrome had been diagnosed. The patients underwent bilateral adrenalectomy as ultima ratio to control life-threatening hypercortisolism after other therapies had failed. Formalin-fixed paraffin-embedded sections were stained as described above. The normal adrenal tissue was derived from adrenal glands removed as part of tumor nephrectomy (n = 6). They were histologically proven normal adrenal glands without neoplastic tissue.

Data of the Emory Predictors of Remission in Depression to Individual and Combined Treatments (PReDICT) (61, 62) study was used to investigate effects of the ABCB1 variant rs2032582 on HPA axis function in 154 unmedicated patients with a current Diagnostic and Statistical Manual of Mental Disorders (DSM)IV diagnosis of major depressive disorder. The PReDICT study was designed and conducted in accord with the latest version of the Declaration of Helsinki. The Emory Institutional Review Board (IRB) and the Grady Hospital Research Oversight Committee gave ethical approval for the study design, procedures, and recruitment strategies (Emory IRB numbers 00024975 and 00004719). The PReDICT study is registered at ClinicalTrials.gov Identifier: NCT03226912 and NCT00360399. DNA was extracted from whole blood, and genome-wide SNP genotyping was performed using HumanOmniExpress BeadChips. Quality control was performed in PLINK. Samples with low genotyping rate (<98%) were removed. SNPs with a high rate of missing data (>2%), significant deviation from the Hardy-Weinberg equilibrium (HWE, P < 105), or a low minor allele frequency (<5%) were excluded from further analyses. SNP genotypes were coded as 0 for major homozygotes (GG, n = 56), 1 for heterozygotes (TG, n = 74), and 2 for minor homozygotes (TT, n = 24) and did not deviate from HWE (2 = 0.27, P = 0.60). HPA axis function was assessed using the dexamethasone/corticotropin-releasing hormone (Dex/CRF) test, consisting of an oral administration of 1.5 mg of Dex at 11:00 p.m. and an infusion of ovine CRF (1 g/kg) at 3:00 p.m. on the next day. Cortisol and ACTH levels were measured from plasma samples taken immediately before CRF administration (pre-CRF) (i.e., at 3:00 p.m.) and again at 3:30 p.m. (30 min), 3:45 p.m. (45 min), 4:00 p.m. (60 min), and 4:15 p.m. (75 min). Baseline cortisol levels were available for all 154 patients with genotype data for the SNP rs2032582. Only completers were included in the analysis, so there are no dropouts in sample sizes over time. Linear regression models were used to test for effects of the SNP genotype on baseline cortisol levels using R version 3.6.2. To assess differences in cortisol and ACTH levels after the Dex/CRF test over time, linear mixed effects models with a random intercept for each patient were applied. All models included gender, age, and baseline depression severity sum scores on the 17-item Hamilton Depression Rating Scale (63) and the first five genetic ancestry (multidimensional scaling) components as covariates.

See the original post here:
Single-cell molecular profiling of all three components of the HPA axis reveals adrenal ABCB1 as a regulator of stress adaptation - Science Advances

Read More...

The Need for New Biological Targets for Therapeutic Intervention in COPD – Pulmonology Advisor

February 1st, 2021 4:45 pm

Chronic obstructive pulmonary disease (COPD) continues to be a major cause of disability and is one of the leading causes of mortality worldwide. While there are numerous treatment options for the lung disease, the available treatments focus on symptoms secondary to inflammation, and are not curative. In a review published in the American Journal of Physiology Lung Cellular and Molecular Physiology, experts focus on potential disease-relevant pathways and emphasize the important of developing new treatments for patients with COPD.1

The objective of the review was to summarize COPD pathology, available treatment options and additional potential pathways and targets for new therapeutic development.

Cigarette smoke contains thousands of injurious agents and is the key cause of COPD worldwide as these induce tissue damage and inflammatory process leading to destruction of alveolar tissue, loss of extracellular matrix and alveolar cells, along with airway remodeling.2 As COPD may progress in patients despite smoking cessation it was suggested that persistent airway inflammation in these patients is related to repair of smoke-induced tissue damage in the airways.3 Failure to achieve normal lung function in early adulthood followed by age-appropriate rates of decline causes up to half of COPD cases.4

The 2020 Global Initiative for Chronic Obstructive Lung Disease guidelines recommend that the management strategy of COPD should be based on the assessment of symptoms and future risk of exacerbations and the main goals of pharmacological therapy for COPD are to reduce symptoms and frequency and severity of exacerbations, as well as to improve exercise tolerance and health status. However, at this point there is no evidence that any of the available medications can modify the long-term decline in lung function.5

The commonly used maintenance medications in COPD are short- and long-acting beta-2 agonists and anti-cholinergics, methylxanthines, inhaled or systemic corticosteroids, phosphodiesterase (PDE)-4 inhibitors and mucolytic agents.5 As these medications are mainly focused on relieving symptoms and reducing the risk for exacerbations, more effective treatment strategies are needed. COPD is a complex disease and precision medicine strategy, that considers biologic and psychosocial factors, may improve disease outcomes.4

New Treatment Targets

There is a real need to uncover new biology in order to advance more precision-based therapeutic strategies for patients with COPD. New disease-specific strategies in development are focusing on inflammatory pathways, hoping this will help to address disease onset. Early reports suggest there are several promising targets that can address inflammatory complications, including oxidative stress, kinase-mediates pathways, phosphodiesterase inhibitors, interleukins and chemokines.

Oxidative Stress a common denominator for aging and cellular senescence, resulting in macromolecular damage and DNA damage.2 With cigarette smoke exposure there is an increased oxidative stress, associated with an increase in Nrf2 activity which declines with the progression of COPD.6 As several studies have implicated Nrf2 in COPD pathology, this pathway is a potential important therapeutic target. Several agents may change Nrf2 expression and activity in airway cell, including aspirin-triggered resolvin D1, crocin, sulforaphane, and schisandrin B.1,6

Kinase-mediated Pathways as various kinases, including MAPK, receptor-tyrosine kinases, phosphoinositide-3-kinases, JAK, and NF-B, may induce chronic inflammation, they may serve as new targets for COPD treatment. There are several drugs that target different kinases but these are not approved for clinical use. Drugs with a more specific action, such as RV568 that inhibits p38, was well tolerated in a 14-day clinical trial and showed promising results with potent anti-inflammatory effects on cell and animal models relevant to COPD, with evidence for improvement in lung function and anti-inflammatory effects on sputum biomarkers.7

Phosphodiesterase Inhibitors inhibiting PDE leads to an increase in intracellular cAMP levels that may have anti-inflammatory effects. Roflumilast is an oral PDE-4 inhibitor already in use for more severe cases of COPD, but more potent medications are being developed, including several inhaled formulations, such as CHF6001, which was reported to have significant anti-inflammatory properties in the lungs of patients with COPD already receiving triple inhaled therapy (8). Ensifentrine is a PDE3/PDE4 inhibitor with anti-inflammatory and bronchodilator properties and when combined with short-acting bronchodilators or tiotropium caused additional improvement in lung function, reduced gas trapping, and improved airway conductance.9

Inflammatory Mediators exposure to inhaled irritants and tobacco smoke results in an increase in various interleukins (IL) that increase the number of immune cells and induce inflammatory responses. Hence, treatment directed against these mediators may reduce inflammation.1 Mepolizumab, reslizumab, and benralizumab are antibodies directed against IL-5 and its receptor and reduced eosinophil-related inflammation. These medications are approved for use for asthma, and were not effective in COPD, but may be valuable for patients with COPD with eosinophilia. Dupilumab, a monoclonal antibody directed against IL-4 and IL-13 receptor, is another potential candidate for future use. microRNAs are also involved in inflammation regulation, and miR-155 expression was shown to be increased in COPD, but at this point there are no available miRNA-based therapeutics for COPD.10

Additional Potential Treatment Targets

While multiple medications under development for COPD are focusing on the inflammatory pathways, they are not expected to reverse the lung damage. For this reason, it is important to study the upstream pathways that may help to identify strategies to reverse exiting lung damage, including targets that can lead to lung repair and regeneration.

These potential breakthrough targets may include treatments directed against mitochondrial dysfunction; structural integrity of airway epithelium such as proteins that comprise tight junctions or the extracellular matrix; various ion channels that are responsible for airway hydration; and pro-regenerative strategies, including stem cell and tissue-engineering treatments to repair lung damage.1

Animal models and 3D human-based disease models have an important role in the efforts to better understand disease process and identify specific therapeutic targets and pathways.11,12 These models improve our knowledge about the basic mechanisms underlying COPD physiology, pathophysiology and treatment. Although they can only mimic some of the features of the disease, they are valuable for further investigation of mechanisms involved in human COPD.11

Several different types of 3D cell culture models have been developed in recent years, and these have gained increasing interest in drug discovery and tissue engineering due to their evident advantages in providing more physiologically relevant information and more predictive data. Ex vivo modeling using primary human material can improve translational research activities by fostering the mechanistic understanding of human lung diseases while reducing animal usage. It is believed that using new model organisms may allow exploring new avenues and treatments approached for human disease, and these are especially promising.12

COPD is a major public health concern, and as it continues to be a global burden, the importance of developing new treatments is apparent. Current treatments are not curative, and while new strategies and drugs are in the pipeline, they still address mostly secondary inflammatory pathways of the disease. An additional major complication in COPD drug development likely comes from the essential dependency on surrogate endpoints like FEV1 to assess the impact of a therapeutic strategy. Thus, any new therapeutic strategy will ultimately require long-term studies to confirm that the surrogate endpoints accurately reflect efficacy on disease outcome, concluded the researchers.

References

1.Nguyen JMK, Robinson DN, Sidhaye VK. Why new biology must be uncovered to advance therapeutic strategies for chronic obstructive pulmonary disease. Am J Physiol Lung Cell Mol Physiol. 2021;320(1):L1-L11. doi:10.1152/ajplung.00367.2020

2.Tuder RM, Petrache I. Pathogenesis of chronic obstructive pulmonary disease. J Clin Invest. 2012;122(8):2749-55. doi:10.1172/JCI60324

3.Willemse BW, ten Hacken NH, Rutgers B, Lesman-Leegte IG, Postma DS, Timens W. Effect of 1-year smoking cessation on airway inflammation in COPD and asymptomatic smokers. Eur Respir J. 2005;26(5):835-45. doi:10.1183/09031936.05.00108904

4.Sidhaye VK, Nishida K, Martinez FJ. Precision medicine in COPD: where are we and where do we need to go? Eur Respir Rev. 2018;27(149):180022. doi:10.1183/16000617.0022-2018

5.Global Initiative for Chronic Obstructive Lung Disease. Global strategy for the diagnosis, management, and prevention of chronic obstructive pulmonary disease 2020 report [Online]. Global Initiative for Chronic Obstructive Lung Disease. https://goldcopd.org/wp-content/uploads/2019/11/GOLD-2020-REPORT-ver1.1wms.pdf. Accessed January 25, 2021.

6.Cuadrado A, Rojo AI, Wells G, et al. Therapeutic targeting of the NRF2 and KEAP1 partnership in chronic diseases. Nat Rev Drug Discov. 2019;18(4):295-317. doi:10.1038/s41573-018-0008-x

7.Charron CE, Russell P, Ito K, et al. RV568, a narrow-spectrum kinase inhibitor with p38 MAPK- and - selectivity, suppresses COPD inflammation. Eur Respir J. 2017;50(4):1700188. doi:10.1183/13993003.00188-2017

8.Singh D, Beeh KM, Colgan B, et al. Effect of the inhaled PDE4 inhibitor CHF6001 on biomarkers of inflammation in COPD. Respir Res. 2019;20(1):180. doi:10.1186/s12931-019-1142-7

9.Singh D, Abbott-Banner K, Bengtsson T, Newman K. The short-term bronchodilator effects of the dual phosphodiesterase 3 and 4 inhibitor RPL554 in COPD. Eur Respir J. 2018;52(5):1801074. doi:10.1183/13993003.01074-2018

10.Barnes PJ. Targeting cytokines to treat asthma and chronic obstructive pulmonary disease. Nat Rev Immunol. 2018;18(7):454-466. doi:10.1038/s41577-018-0006-6

11.Ghorani V, Boskabady MH, Khazdair MR, Kianmeher M. Experimental animal models for COPD: a methodological review. Tob Induc Dis. 2017;15:25. doi:10.1186/s12971-017-0130-2

12.Zscheppang K, Berg J, Hedtrich S, et al. Human pulmonary 3D models For translational research. Biotechnol J. 2018;13(1):1700341. doi:10.1002/biot.201700341

Continue reading here:
The Need for New Biological Targets for Therapeutic Intervention in COPD - Pulmonology Advisor

Read More...

IDEXX and the Tuskegee University College of Veterinary Medicine Announce Landmark Initiative – Conway Daily Sun

January 31st, 2021 2:51 am

Advancing diversity in veterinary medicine through nine full scholarships and integrated support

WESTBROOK, Maine, Jan. 28, 2021 /PRNewswire/ -- IDEXX Laboratories, Inc. (NASDAQ: IDXX), a global leader in veterinary diagnostics and software, announced today a landmark initiative with the Tuskegee University College of Veterinary Medicine (TUCVM), which it intends to support by a contribution of $3.6 million over six years from the IDEXX Foundation, a donor-advised fund administered by a national donor-advised fund program. This intended support represents the IDEXX Foundation's inaugural investment as part of a larger effort to advance diversity, equity, and inclusion in veterinary medicine.

The TUCVM is the only veterinary medical professional program in the United States located on the campus of a Historically Black College or University (HBCU). A pioneer in veterinary medical education for 75 years, the TUCVM has a proven track record of increasing access to veterinary medical education, educating 70% of African American veterinarians.

"We are honored to support the Tuskegee University College of Veterinary Medicine with an investment from the IDEXX Foundation, designed to ensure positive student outcomes through integrated support across the student experience," said Jay Mazelsky, IDEXX President and Chief Executive Officer. "The past 10 years have seen tremendous growth in the diversity of the pet-owning population. Ensuring all pets have access to the highest standard of care in their communities starts with advancing diversity, equity, and inclusion in veterinary medicine."

The TUCVM initiative is designed to drive comprehensive and positive outcomes for diversifying the veterinary field. The initiative includes nine full scholarships, mental health support for veterinary students, emergency funding for students in need, and monies for important capital improvements at the TUCVM facilities. Each of these elements was identified through engagement with the TUCVM leadership to maximize the positive impacts of this meaningful collaboration.

"This is the most impactful contribution that our beloved college has received in our 75 years of existence and recognizes our legacy of work training and educating students of color,"said Ruby L. Perry, DVM, MS, PhD, Diplomate-ACVR, Dean of the Tuskegee University College of Veterinary Medicine. "This meaningful investment by the IDEXX Foundation will help our students realize their dreams of becoming veterinarians."

About the IDEXX Foundation

The IDEXX Foundation is a donor-advised fund administered by a national donor-advised fund program. The IDEXX Foundation's mission is to create positive, lasting impacts for people, animals, and the environment through inclusive and outcomes-focused initiatives in communities around the world. In addition to creating opportunities for the diverse leaders of tomorrow, the IDEXX Foundation also prioritizes projects that improve access to veterinary care for underserved communities, disaster relief, and animal-assisted therapy, and cultivates community-focused solutions to the global challenges of access to safe water, impacts of climate change, and food security.

Corporate Responsibility at IDEXX

Corporate responsibility is fundamental to how IDEXX operates as a business, employer, supplier, customer, and member of the community. The company prioritizes investments that support its Purpose and Guiding Principles and is committed to the highest ethical standards, empowering and rewarding employees, promoting a culture that values diversity, equity, and inclusion, and seeking to enhance environmental sustainability in its facilities and operations.

About IDEXX Laboratories, Inc.

IDEXX Laboratories, Inc. is a member of the S&P 500 Index and is a leader in pet healthcare innovation, offering diagnostic and software products and services that deliver solutions and insights to practicing veterinarians around the world. IDEXX products enhance the ability of veterinarians to provide advanced medical care, improve staff efficiency, and build more economically successful practices. IDEXX is also a worldwide leader in providing diagnostic tests and information for livestock and poultry and tests for the quality and safety of water and milk and point-of-care and laboratory diagnostics for human medicine. Headquartered in Maine, IDEXX employs more than 9,000 people and offers products to customers in over 175 countries. For more information about IDEXX, visit idexx.com.

About the Tuskegee University College of Veterinary Medicine

Located in Alabama as one of the state's two accredited veterinary programs, it was envisioned in 1944 by Dr. Frederick D. Patterson, founder of the United Negro College Fund (UNCF), and officially established at Tuskegee in 1945. It is the only veterinary medical professional program located on the campus of a Historically Black College or University (HBCU) in the United States. The College's primary mission is to provide an environment that fosters a spirit of active, independent, and self-directed learning, intellectual curiosity, creativity, critical thinking, problem-solving, ethics, and leadership; and promotes teaching, research, and service in veterinary medicine and related disciplines.For more information, visittuskegee.edu/vetmed.

Read this article:
IDEXX and the Tuskegee University College of Veterinary Medicine Announce Landmark Initiative - Conway Daily Sun

Read More...

The role of cannabis in veterinary medicine – Vet Candy

January 31st, 2021 2:51 am

A new scientific review finds relevant data from cannabis medical studies in companion animals. Nowadays, both low-THC and high-THC cannabis are used for medical uses. However, the majority of veterinary cannabis products contained low-THC.

The endocannabinoid system (ECS) was characterized and described as a complex regulatory system that provides essential homeostasis mechanisms through the body. This system has been studied in humans and several animals, from birds to canines. It also plays an important role and is divided into three components: cannabinoid receptors (CBRs), endocannabinoids (eCBs), and enzymes responsible for the activation, transportation, and breakdown of eCBs.

Most of the veterinary cannabis products have been clinically used for osteoarthritis and epilepsy in canines. For these patients, clinical research has suggested doses of 2 mg/k. The pain assessment scales have decreased after a few weeks of treatment with CBD oil, but there have not been reported changes in lameness.

Previous studies have shown that products with nearly little or no THC had efficacy to treat seizures, mild pain, and anxiety. Also, another study with oral and transdermal CBD reported being well-tolerated in canines. Interestingly, it has been reported that feeding might enhance CBD absorption compared to animals without feeding. Besides, other study finds that the combination of CBD dominant oil with standard anticonvulsant therapies has reduced the seizure frequency of canines.

But, there is still a limited amount of research data regarding the safety and use of cannabis in veterinary medicine. It has been advised to veterinarians to apply their use carefully and consult their state veterinary medical board.

For that, it's essential to properly understand each Cannabis compound's mechanism to develop a better clinical approach or treatments.

To read more click on the link below:

Cannabis in Veterinary Medicine: A Critical Review

Read the rest here:
The role of cannabis in veterinary medicine - Vet Candy

Read More...

New Year Begins with Special Emphasis on Diversity and Inclusion in PVM – Purdue Veterinary News

January 31st, 2021 2:51 am

Friday, January 29, 2021

During the week surrounding Martin Luther King Jr. Day, the College of Veterinary Medicines chapter of VOICE (Veterinarians as One Inclusive Community for Empowerment) hosted Inclusive Excellence Week with several virtual events focused on the theme, Finding Our Voices. VOICE provides a common ground for veterinary students, staff, and faculty from all backgrounds to exchange ideas and celebrate diversity.

As a minority entering a predominantly white career field, organizations like Veterinarians as One Inclusive Community for Empowerment have been major influencers in creating a sense of inclusion and hope for me and other students, said Malaycia Goldsmith, president of Purdue VOICE. There is power in actions of inclusion. That power is formed by making connections, learning from, and uplifting people that have differences actively working to listen, understand, and support one another. Malaycia further shared, As a member and the president of our Purdue VOICE chapter, I have continuously been filled with gratitude to be surrounded by and learn from the people within this community, whether they are our members or supporters. I hope that our organization can continue to shed light on socio-cultural issues and influence all of us to continue to reflect and see how we can play a part in increasing diversity in spaces that need it.

Kicking-off Inclusive Excellence Week, the MLK Day of Service included a food drive in support of Purdues ACE Campus Food Pantry. Food donations were accepted all week long in the colleges Office for Diversity, Equity, and Inclusion in Lynn Hall as well as at locations around campus.

A Q&A Panel entitled, Making Room in Veterinary Medicine, featuring Danielle Lambert and Dr. Tierra Price, was hosted via Zoom on Tuesday, January 19. Danielle Lambert is the founder of Snout School, an organization dedicated to supporting women in veterinary medicine. Dr. Tierra Price is the founder of the Black DVM Network, an organization for Black veterinary professionals to connect and grow while increasing the exposure of diversity in veterinary medicine.

A virtual panel discussion on Wednesday, January 20, featured some familiar faces to the PVM family. The panel consisted of Dr. Henry Green, the first African American veterinary cardiologist and a former PVM faculty member; Dr. Margie Lee, one of the first African American veterinary graduates of the Virginia Maryland College of Veterinary Medicine; and PVM alumna, Dr. Mary Beth Leininger (PU DVM 67), the first woman to serve as president of the American Veterinary Medical Association. The trio led the panel discussion on Breaking the Ceiling: Significant Firsts in Veterinary Medicine.

The Inclusive Excellence Week schedule continued Thursday, January 21 with a Zoom session featuring Dr. Kate Toyer, the president of the Australian Rainbow Veterinarians and Allies, an organization dedicated to supporting Australian Veterinarians who are part of the LGBT+ community. Dr. Toyer led the open forum focused on creating the new norm in veterinary medicine.

Finally, on Friday, January 22, the week of activities was capped-off with a virtual Amazon Prime Watch Party for the movie Hidden Figures, which tells the story of a team of female African American mathematicians who played a pivotal role in NASAs success during the Space Race in the 1960s. VOICE put a lot of work and effort into Inclusive Excellence Week, and it showed, said Dr. Latonia Craig, the colleges assistant dean for inclusive excellence. I was happy to see so many faculty, staff, and students support their programming. We want to do everything in our power to continue to promote engagement in the area of diversity. Im so proud that VOICE is an extension of the PVM Office of Diversity, Equity, and Inclusion. Their commitment and innovative programming will take this college to greater heights.

Writer(s): Jonathan Martz, PVM Communications Intern | pvmnews@purdue.edu

See the original post:
New Year Begins with Special Emphasis on Diversity and Inclusion in PVM - Purdue Veterinary News

Read More...

Mistrust May Be Why Some Dog Owners Avoid The Vet – The Bark

January 31st, 2021 2:51 am

When it comes to veterinary care, barriers to access, including a lack of trust on the part of dog owners, play a bigger role than differences in race, gender, or socioeconomic status, according to a new study. Findings could help veterinarians develop outreach strategies for underserved communities.

I was interested in how different demographic groups viewed health care and how those views might affect relationships between veterinarians and their clients, says study first author Rachel Park, a PhD student at North Carolina State University. The existing literature wasnt national in scope and hadnt accounted for multiple identities held, such as ones socioeconomic status or education, so I saw a knowledge gap that could be filled.

For the study inVeterinary Sciences, Park used Amazons Mechanical Turk to conduct an online survey of 858 self-identified dog owners. The survey asked participants to indicate how likely they would be to seek veterinary care under 18 different circumstances. The survey also asked participants supplemental questions about their relationship with their dog, previous veterinary behavior, and demographic information.

While there was some variance for different medical scenarios, the overall likelihood of dog owners to seek care did not differ significantly across demographics, regardless of race, gender, or socioeconomic status. However, there were demographic differences related to barriers to veterinary care, as well as to the owners relationship with the dog.

Sign up for our newsletter and stay in the know.

We did see that women (58.0%), white (48.9%) and Asian (64.4%) dog owners were more likely to describe the dog as a member of the family, rather than as property, whereas some Black (24.4%) or Native American (25.0%) dog owners were more likely to consider the dog property, says Park. But the difference in the way therelationshipwas described didnt carry over into a difference in likelihood to seek veterinary care.

The primary barriers to care that respondents identified were transportation, veterinary office hours of operation, cost, language differences, and trust. Cost was a bigger factor for dog owners under 29 years old or households making less than $60,000 per year. However, these barrierswith the exception of trustcut across all demographics: race, gender, education level, and socioeconomic status.

Black and Native American respondents were about 10 to 15% more likely to indicate a lack oftrustas a barrier to seeking veterinary care.

This was the most interesting finding, Park says. Respondents had the ability to select had a bad prior experience with veterinarian as an option, but those who indicated lack of trust didnt choose that as the reason. Research has long reported that racial and ethnic minorities often experiencemistrustin health professionals in human medicine and consequently seek health care at a lower rate.

Our findings reveal that Black and Native American dog owners experience a similar mistrust in veterinary medicine. This appears to be an important avenue for future research.

While there are limitations to the surveyno statistical weighting to adjust for over- or under-samplingPark says the results are still useful for identifying opportunities for outreach from the veterinary community.

Im hopeful that this study can help us better understand the barriers different communities face, Park says. Everyone wants to do whats best for theirdog, so the veterinary community has the opportunity to help ensure equal access to care and try and ease those barriers.

Original post:
Mistrust May Be Why Some Dog Owners Avoid The Vet - The Bark

Read More...

Wolf Administration Awards $1.3 Million to Fund Research to Grow PA Agriculture Industry – Governor Tom Wolf

January 31st, 2021 2:51 am

Harrisburg, PA - Agriculture Secretary Russell Redding today announced grants totaling $1.287 million to eight organizations for research on issues critical to sustaining and growing Pennsylvanias agriculture industry. Grant recipients include Pennsylvania State University, Temple University, University of Pennsylvania School of Veterinary Medicine, American Mushroom Institute, Baarda Farms, Coexist Build, Pasa Sustainable Agriculture and Team Ag, Inc.

Meeting the challenges of feeding a growing population amid rapid changes in climate, technology and animal and plant diseases demands investment in research and development, said Redding. These investments hold the promise and potential to spur the innovation we need to increase productivity; advance human and animal medicine; and support cleaner water, healthier soil and a safer food supply.

The grants, awarded by the Pennsylvania Department of Agriculture, focus on a broad range of research topics including detecting COVID-19 exposure in livestock, increasing farm productivity and profits, protecting pollinators, safely controlling Spotted Lanternfly and other invasive species and improving soil and water quality and sustainability through regenerative farming.

This funding supplements $900,000 in agricultural research support through the departments budget to Rodale Institute, the Penn State University Center for Agricultural Law, Penn State Extension, and the Centers for Beef, Dairy, Poultry and Livestock Excellence.

Following is a list of 27 grantees, amounts awarded and project titles:

MEDIA CONTACT: Shannon Powers - 717.603.2056, shpowers@pa.gov

# # #

Originally posted here:
Wolf Administration Awards $1.3 Million to Fund Research to Grow PA Agriculture Industry - Governor Tom Wolf

Read More...

Honor the Veterinary Heroes In Your Animal’s Life – PRNewswire – PRNewswire

January 31st, 2021 2:51 am

American Humane, the country's first national humane organization, recognizes the hard work these professionals put in day-in and day-out in the name of helping animals, and is pleased to announce that nominations are now open for the eighth annual American Humane Hero Veterinarian and Hero Veterinary Nurse Awards, presented by Zoetis Petcare (a U.S. business unit of Zoetis). Pet owners and animal lovers alike are invited to visit http://www.herovetawards.org between now and March 24 to nominate their favorite veterinary professionals dedicated to the betterment of the health and welfare of animals and the promotion of the people-animal bond. The winning veterinarian and veterinary nurse will be featured in the 11th annual American Humane Hero Dog Awards broadcast this fall, which will air nationwide as a two-hour special on Hallmark Channel.

These awards are not limited to companion animal veterinarians. Professionals from all fields of veterinary medicine are eligible for entry including, but not limited to those who work in research, emergency services, shelters, and those who work with large and exotic animals. The five finalists in the American Hero Veterinarian and American Hero Veterinary Nurse categories will be selected by a special blue-ribbon panel of judges consisting of veterinary professionals, animal care professionals and celebrities. Beginning June 10, the American public will be invited to vote online for their favorite veterinarian and veterinary nurse.

"Zoetis is honored to support America's veterinary community, and the American Humane Hero Veterinarian and Hero Veterinary Nurse Awards are our way of thanking these devoted individuals," said Tara Bidgood, DVM, PhD, DACVCP, executive director, Zoetis Petcare Veterinary Professional Services & Medical Affairs. "Veterinarians and veterinary nurses are leading the effort to keep America's animals happy and healthy, and they deserve recognition from a grateful nation."

"The world's animals depend on veterinarians and veterinary nurses, and we thank Zoetis Petcare, the exclusive sponsor of the Hero Veterinarian and Hero Veterinary Nurse Awards, for helping recognize their achievements in saving lives, conducting groundbreaking research, and working on the front lines of animal welfare," said American Humane President and CEO Dr. Robin Ganzert. "To us, all vets and vet nurses are heroes and these awards help us shine a bright spotlight on the best of the best."

Key dates for the 2021 American Humane Hero Veterinarian and Hero Veterinary Nurse Awards contest are:

Nominations Period:

Jan. 27 March 24

Public Voting Round:

June 10 July 29

Hero Dog Awards broadcast:

Coming this fall

*All rounds open and close at noon Pacific Time

To nominate a veterinarian or veterinary nurse between now and March 24, and for complete contest rules, please visit http://www.herovetawards.org. To nominate a Hero Dog in your life for the 2021 American Humane Hero Dog Awards, please visit http://www.herodogawards.org between now and March 2.

About American HumaneAmerican Humane is the country's first national humane organization. For more information please visit http://www.americanhumane.org, and please follow us on Facebook and Twitter.

About ZoetisZoetisis the leading animal health company, dedicated to supporting its customers and their businesses. Building on more than 65 years of experience in animal health, Zoetis discovers, develops, manufactures and commercializes medicines, vaccines and diagnostic products, which are complemented by biodevices, genetic tests and precision livestock farming. Zoetis serves veterinarians, livestock producers and people who raise and care for farm and companion animals with sales of its products in more than 100 countries. In 2019, the company generated annual revenue of $6.3 billion with approximately 10,600 employees. For more information, visit http://www.zoetis.com.

SOURCE American Humane

Home

See original here:
Honor the Veterinary Heroes In Your Animal's Life - PRNewswire - PRNewswire

Read More...

MSU veterinary imaging research aims to help both man and his best friend – The Reflector online

January 31st, 2021 2:51 am

Mississippi State University's College of Veterinary Medicine is working to advance early disease detection in animals through their ground-breaking research in imaging technologies.

According to Dr. Alison Lee, doctor of veterinary medicine and assistant professor at MSU, the Department of Veterinary Medicine is using imaging technology such as CT and MRI machines to detect issues like brain tumors and inflammatory and infectious diseases in an animal's brain.

Lee said this research is conducted at an imaging center on Stark Road. Researchers are primarily using the MRI machine to focus on a brain tumor study.

"The MRI allows us to see central nervous tissuethe brain and the spinal cordmuch better than any other imaging technologies let us see it. It can tell us when there is anything abnormal in the brain," Lee said.

Dr. Andy Shores, clinical professor and CVM chief of neurosurgery and neurology, said this research is profound because brain tumors affect humans and animals in very similar ways, leading to advancement for the treatment of both groups.

Shores said the National Institute of Health has provided funding for MSU's researchers to look at novel ways to treat brain tumors, specifically glioblastoma, in both humans and animals.

"The dog is the model for the human disease because there are so many similarities with the type of tumor and the way it affects them," Shores said.

The neurosurgery neurology group that is focusing on imaging technologies is not merely performing research but instead applying it to real-life diagnosis and treatments.

"A lot of what we do is not really research but clinical activity," Shores said. "We incorporate what we are able to do with the patients and further the advancement of treatment for certain diseases."

According to Shores, he takes dogs who have brain tumors and uses the MRI to diagnose and plan for a surgical removal of the brain tumor. The dogs receive an injection of a modified virus designed to attack only tumor cells.

Shores then performs follow-up routine exams and imaging to check on the dog. Additionally, he puts them on another drug which helps to uncover the tumor and attack it.

Lee helps Shores assess the MRI images and decide which tumor is likely. She said they can use ultrasound imaging technology to help further localize the tumor during a surgery.

Imaging technology is helpful to use not only during surgery but also during follow-up treatment. Additionally, Lee helps Shores use cross-sectional imaging during the post-operative stage.

Lee is passionate about imaging technologies because brain tumors are devastating in both species. She said they cause behavioral issues and affect everything from an animal's ability to eat and drink to their ability to urinate.

"The brain is a difficult area to treat because we do not have a great understanding of exactly how it works and it is also covered by the skull, so it is a difficult area to operate on," Lee said. "All of what we are doing research-wise is going to help us better treat these animals and lead to longer survival times for both people and animals."

Bailey Haller, a senior biological sciences major from Gulfport, said MSU's research is extremely important for not just the veterinary medical field but the medical field as a whole.

"Mississippi State is finding ways to help animals that previously would not have been possible," Haller said. "They are also developing equipment and research that will be further explored for human use."

Haller decided to be pre-vet because she loves helping animals and learning how they function. She said she is honored to be a part of the program and hopes to work on research like this in the future.

Chief of Neurosurgery and Neurology Shores believes research like this is important because most people view their animals as companions, and that has made it increasingly important to make sure animals can maintain their health.

"Animals have evolved from being a pet around the house to being actual companions, family members and emotional support," Shores said. "Being able to look at those kinds of diseases and further treatment is a benefit to the animal and the human population because of their emotional attachment to their animals."

Lee hopes people are aware MSU has this technology and that this type of technology is not everywhere. She said if anyone has concerns about their pet, MSU's Animal Health Center is a wonderful place to visit.

"We can certainly help them get diagnoses and figure out the next best step for treatment," Lee said. "We are very lucky here to be able to offer this type of technology to clients."

Read more:
MSU veterinary imaging research aims to help both man and his best friend - The Reflector online

Read More...

Presentation Highlights Oncology Research Success | Purdue University College of Veterinary Medicine – Purdue Veterinary News

January 31st, 2021 2:51 am

Tuesday, January 26, 2021

Distinguished Professor of Comparative Oncology and Dolores L. McCall Professor of Comparative Oncology Deborah Knapp recently marked a milestone in her career. As an endowed professor, Purdue University requires Dr. Knapp undergo a review by her department every five years with the latest review completed last month. Dr. Knapp was first named the Dolores L. McCall Professor of Comparative Oncology in 2006 and has held the title ever since. As part of the review, Dr. Knapp was asked to present a seminar highlighting some of her research teams success.

The seminar entitled, Cancer Prevention in Dogs: Strategies That Can Be Implemented Now to Improve Outcomes, was presented virtually December 11, 2020. After an introduction by Dr. Catharine Scott-Moncrieff, head of the PVM Department of Veterinary Clinical Sciences, Dr. Knapp began her lecture by describing the current state of cancer cases in the United States. Last year, there were 1.7 million new cases of human cancer reported with 600,000 of these resulting in death. As for dogs, approximately 4 million cases were expected in 2020 with the total number of deaths unknown. Dr. Knapps team has tasked themselves with looking at ways to improve the outcome for humans and dogs.

In describing a key to improving the outcome for both, Dr. Knapp explained that certain forms of naturally-occurring cancer in dogs are very closely related to cancers in humans, allowing treatment methods to be studied interchangeably. In addition to defining cancer by the organ in which it originates (i.e. breast cancer), researchers have found that in many cases they can better define the cancer by its mutations and molecular makeup of the cancer. For instance, a subset of bladder cancer in dogs may be more closely related to colon cancer in people.

Dr. Knapp described that one way to improve cancer outcomes in humans and dogs is to improve the use of current drugs such as Cyclooxygenase (Cox) Inhibitors, also known as Non-Steroidal Anti-Inflammatory Drugs. These drugs have anti-cancer properties and are a good example of drugs that can be repurposed. Additionally, new drugs, such as immunotherapies, are currently being studied. The Purdue University College of Veterinary Medicine has recently received a research grant to improve knowledge of these methods. Another area that has the potential to make a big difference in cancer outcomes for humans and dogs is individualized care.

The primary focus of improvement described in the lecture is prevention. Cancer prevention comes in three forms. Primary cancer prevention is preventing the entire cancer development, such as not smoking to avoid lung cancer. Secondary cancer prevention is the detection of precancerous symptoms with testing such as mammograms and colonoscopies and treating them before they become aggressive cancers. Tertiary cancer prevention is the treatment of cancer once it is diagnosed to prevent morbidity and mortality.

Dr. Knapp focused on prevention in regard to bladder cancer, which most of the time is known as high grade invasive urothelial carcinoma, transitional cell carcinoma, aggressive bladder cancer, or in humans is called muscle invasive bladder cancer all essentially the same disease. Dr. Knapp said primary cancer prevention for dogs includes limiting risk factors such as the use of lawn care chemicals; obesity; the use of old generation flea, tick, and mange dips; and exposure to second-hand smoke. Emerging data now suggest that exposure to smoking could be important to bladder cancer in dogs. On the other hand, feeding vegetables to dogs can reduce the risk of cancer. There is now evidence to suggest spayed and neutered dogs are more at risk for cancer than dogs that are not spayed or neutered. Dr. Knapp certainly does not recommend ending the practice of spaying and neutering entirely, but said the optimal time for surgery requires further study, and this raises the question of whether waiting until the dog is a year old should be considered. More studies are needed to answer this question.

Secondary cancer prevention of bladder cancer can come in the form of early detection of precancerous lesions such as dysplasia and carcinoma in situ. In this stage, the cancer is less advanced and should be easier for the drugs to treat. Additionally, the immune system should be in a more active state to combat the cancer. This prevention strategy was tested by Dr. Knapp and her team in an early detection, early intervention study in which Scottish Terriers, a high-risk breed for bladder cancer, were studied. The study was completed in collaboration with the Scottish Terrier Club of America, which provided funding. The dogs were examined every six months for three years. Preliminary study results show that bladder cancer can be detected early, and early detection does improve the outlook for the dogs. Further evaluation is currently underway by Dr. Knapp and her study collaborators.

There are several key strategies that can be implemented now to improve cancer outcomes. Avoiding factors that increase the bladder cancer risk is important. Early detection and intervention are emerging as vital to improving the outcome for dogs with bladder cancer.

Dr. Knapp earned her DVM degree at Auburn University and in 1985 came to Purdue where she completed her residency and earned a masters degree in 1988. She then became board certified by the American College of Veterinary Internal Medicine (Oncology). She was named a Distinguished Professor of Comparative Oncology by the university in 2020. Dr. Knapp leads the Purdue Comparative Oncology Program and is regarded as the leading expert in the field of naturally occurring bladder cancer in dogs. Her research has established bladder cancer in dogs as the most relevant animal model for invasive bladder cancer in humans. She is the author of more than 100 peer-reviewed journal articles related to the subject.

Writer(s): Jonathan Martz, PVM Communications Intern, and Allison Carey | pvmnews@purdue.edu

Continued here:
Presentation Highlights Oncology Research Success | Purdue University College of Veterinary Medicine - Purdue Veterinary News

Read More...

On the road with the traveling veterinary nurse, Lucy Taylor – Vet Candy

January 31st, 2021 2:51 am

Most veterinary nurses finish school and find a clinic to work in for the rest of their lives. Although they may travel for pleasure, their work is relatively stationary, so customers know where to find the high quality vet care they need.

At first, Lucy thought she would be following a similar path, but a school trip abroad changed everything for her. She took an Erasmus trip to Estonia, and volunteered at an animal shelter there. The trip changed her. She took another trip, this time with the focus of conservation in South Africa the following year, and knew what she wanted to do with her life.

Lucy made a bold move after veterinary school, and became a traveling locum nurse. She has since been to Spain, Sardinia, and Morocco for her work. While she is in these locations, she often volunteers with organizations such as Vets Beyond Borders, so she can help make a difference in the lives of animals in need.

A typical volunteer day may involve working closely with vets to examine, anesthetize, and spay or neuter a long string of dogs as quickly as possible. Not only are the animals spayed or neutered, but other procedures that may benefit the animal are done at the same time while they are under. This could mean extractions to remove decaying teeth, or enucleations.

Sometimes the animal is too unhealthy for surgery, at which point they would call and recommend to the shelter or who ever they are currently helping that the animal not undergo surgery. Other complications, such as retained testicles, can also make it harder to do the surgery.

Often, the veterinary team ends up missing lunch or working late into the night, in order to help as many animals as possible. While it is difficult work, they are happy to do it because that means one more animal is getting the help it needs.

Lucy is passionate about her job, and loves every moment of helping animals while she travels the world. While she knows this job is the perfect one for her without a doubt, if she hadnt become a vet she did have other aspirations to follow.

Her original childhood dream was to become a zookeeper, which she might have pursued if veterinary school had not have been an option. Her other choice would be as an entertainer. In fact, she still sometimes plays an Elf at Christmas time in Lapland, Finland!

When shes not an Elf, she still finds the ability to entertain but also educate through a podcast about becoming a traveling veterinary nurse she does with co-host Amber LaRock. In it they talk about traveling veterinary medicine, and the unique aspects of trying to help animals while on the road.

Lucy enjoys every minute of her life as a traveling veterinary nurse. She has helped save thousands of needy dogs through her efforts in far away countries, and has also been an inspiration for veterinary students who dream of helping animals, but also traveling too.

Here is the original post:
On the road with the traveling veterinary nurse, Lucy Taylor - Vet Candy

Read More...

IMMS and MSU veterinarians work to save dolphin – WXXV News 25

January 31st, 2021 2:51 am

So far, so good but researchers and care takers at the Institute for Marine Mammal Studies, as well as workers with Mississippi State Universitys College of Veterinary Medicine have their hands full caring for a young male dolphin they rescued Sunday.

This young dolphin fell into the right hands and has a second lease on life thanks to good Samaritans who called the IMMS Sunday morning to report the young juvenile male dolphin appeared to be struggling in the shallow waters of the Mississippi Sound in Gulfport.

Once here at the Gulfport facility, workers with the IMMS and MSUs College of Veterinary Medicine did blood work and diagnostics to assess the state of his condition. His care spilled over in to Monday. Clinical Instructor and Veterinarian at MSU College of Veterinary Medicine Christa Barrett said, Hes still not strong enough to keep himself up, so we have had people in the water here 24-7, and staff here 24-7 with him, to make sure he is able to breathe. Right now, we have Theresa in the water to make sure he is able to breathe. We gave him a combination of some milk, vitamins and things like that to help keep up with his nutritional status.

His mother was nowhere to be found. He does not appear to yet be weaned, but fortunately for this little guy, hes now in capable hands and its not sink or swim time just yet. Hes still in very critical condition, but were doing everything we can. We also have a great faculty staff at Mississippi State University in case we need any specialists to weigh in on the case as well.

As in this case, the IMMS asks anyone who sees a stranded dolphin or sea turtle to contact them as soon as possible. IMMS Director Dr. Moby Solangi said, Stranding season is coming up. Its very important if people see a sick or injured dolphin to call IMMS at 888-SOS-DOLPHIN.

In the meantime, this dolphin will continue to receive critical and supportive care to get him to swim on his own again and hopefully be released back into the Mississippi Sound.

More here:
IMMS and MSU veterinarians work to save dolphin - WXXV News 25

Read More...

This wildlife vet tracks deadly microbes in the African jungle. Now, he’s on the trail of COVID-19 – Science Magazine

January 31st, 2021 2:51 am

Fabian Leendertz has spent decades studying how diseases flow between humans and wildlife. Here, he and colleagues hunt for bats in Ivory Coast.

By Kai KupferschmidtJan. 28, 2021 , 12:05 PM

TA NATIONAL PARK IN IVORY COASTThe message arrived as Fabian Leendertz was watching what he calls breakfast TV: a troop of black-and-white colobus monkeys leaping acrobatically through the trees that tower above the remote field camp here near the Liberian border. A colleague had received word that the carcass of a duiker, a kind of antelope, had been spotted in the rainforest some 10 kilometers away.

The notification launched Leendertz, a wildlife veterinarian at the Robert Koch Institute, into a race against time. The jungle is a hungry place, and Leendertz and his team needed to hike to the carcass before it was hauled away by a leopard or consumed by smaller animals. If the researchers beat the scavengers, they could collect tissue and other materialsincluding maggots feeding on the carrionthat could help answer a fundamental question: What do animals in the jungle die of?

Leendertz and his colleagues have been chasing answers here in the Ta forest for the past 20 years, studying hundreds of carcasses and sampling living animals in one of the only long-term studies of its kind. Theyve found that poachers and predators arent the only deadly threat lurking in the rainforestinfectious diseases are a big killer, too.

A discovery that leprosy can infect wild chimpanzees broke new ground.

The findings have implications for both saving endangered animals, especially apes, and protecting human health. Leendertzs work has revealed, for example, that chimpanzees can die from common cold viruses introduced by humans, prompting scientists, conservation groups, and ecotourism firms to impose new requirements on people visiting the apes. His team has also discovered a previously unknown variant of anthrax that appears to pose a major threat to wildlife. And he and colleagues in Guinea-Bissau recently found that wild chimpanzees suffer from leprosy, suggesting apes might be a previously undetected reservoir of that disfiguring disease, which could spill over into human populations. Fabians work has really changed how we view biosafety and biosecurity around great apes in the wild, says disease ecologist Tony Goldberg of the University of Wisconsin, Madison.

Now, the 48-year-old Leendertz, who once investigated the animal origins of an Ebola outbreak in West Africa, has been asked to help to solve one of the great disease mysteries of the early 21st century: the origins of SARS-CoV-2, the coronavirus that originated in bats and has killed more than 2 million people worldwide. In November 2020, the World Health Organization (WHO) named him to a 10-person team that is examining how the COVID-19 pandemic emerged. At the same time, Leendertz is worrying about how the coronavirus might affect great apes if it spreads to those vulnerable species.

Just 30 minutesafter the message arrived, Leendertz and two other veterinarians, Penelope Carlier and Bernard Ngbocho Nguessan, set off to find the carcass. After a kilometre or so, they passed a group of sooty mangabey monkeys lounging on logs. The animals, even a mother hugging a baby to her belly, appeared undisturbed by the hikers. That is because the monkeys had been habituated; researchers followed them for years until they grew used to humans.

In 1979, primatologists Christophe Boesch and Hedwige Boesch-Achermann came to the forest, one of the last large swaths of rainforest in West Africa, to study chimpanzee behavior. Over years, they habituated chimps, mangabeys, and several other kinds of monkeys, and began documenting their lives. But then, in 1994, the chimpanzees started to die. Eight of 43 study animals turned up dead; four more disappeared.

Before necropsying wild animals, Fabian Leendertz and Kouadio Leonce don protective gear.

The researchers hauled one chimps body back to their camps sturdy dining table for dissection. They wore gloves, but no gowns or masks, and 1 week later one woman fell ill. She recovered, but scientists isolated a virus from her blood. It was a new species of Ebola, a group of viruses already known from human outbreaks elsewhere in Africa, and the dead chimp carried it, too. The discovery of what became known as Ta forest Ebola marked the first time an Ebola outbreak had been documented in nature.

The experience was a wake-up call from both a safety and a scientific perspective, says Boesch, who retired as director of the Max Planck Institute for Evolutionary Anthropology in 2019. In retrospect it is clear that we ran a risk; we were not prepared at all, we had no equipment. And it made the researchers realize that infectious diseases could be playing a larger role in wildlife mortality than they realized. We cannot go on like this, Boesch recalls thinking. They needed a trained veterinarian, and in 2001 Leendertz got the job.

It was the kind of position he had long coveted. Growing up in Krefeld, Germany, Leendertz had bred mice and toads and spent a lot of time at the local zoo. (The director was a friend of his parents.) At university, he began to study biology but grew frustrated. It was just way too much biochemistry, he recalls, including countless hours in the laboratory running polymerase chain reactions (PCRs) to amplify snippets of DNA. All these PCRs were so far removed from working with actual animals, he says, so he switched to veterinary medicine.

In 1999, after completing his undergraduate studies and working in Namibia for a few months, Leendertz reached out to Boesch, asking whether he could join the Ta project. The reply was yesif Leendertz found an academic laboratory that would help support his graduate studies.

That wasnt easy. But Beatrice Hahn, a virologist at the University of Pennsylvania, had just published work showing HIV, the virus that causes AIDS in humans, had come from chimpanzees. The discovery ignited scientific interest in zoonoses, diseases that jump from animals to humans. That was kind of the first big aha! moment about zoonotic disease, Goldberg says. It helped Leendertz find a home at the Robert Koch Institute and ensured that, from the start, he would focus on both veterinary and human medicine.

Researchers draw blood from a dog in a village in Ivory Coast as part of their efforts understand how pathogens move among species.

Starting in 2001, Leendertz spent 14 months at Ta, following chimpanzees through the forest, collecting feces, and conducting necropsies. That was the real starting point for my work, he says. The conditions didnt bother him. He was content to be outdoors and largely cut off from the world, able to send and receive emails just once a week though a satellite connection. Leendertz didnt see images of the planes hitting New York Citys twin towers in 2001 until the year after the attack, after he emerged from his sojourn in the forest.

After a long march,the team found what was left of the dead duiker, surrounded by buzzing flies. Leendertz and Carlier suited up: masks, body suits, face shields, layers of gloves. They filled a bucket of bleach to disinfect equipment. Then they began cutting snippets of tissue and collecting blood, even grabbing a few maggots, which would be liquefied and analyzed for any pathogens they carried.

Moving deliberately around the forest in their gleaming white suits, the researchers looked like investigators at a crime scene. They were, in a way, with the added complication that the killer might still be lurking nearby. Leendertz has been following one suspect in particular since his first stint in the Ta forest. He was watching a group of chimpanzees when an alpha male named Leo suddenly vomited. Then, He climbed on this low branch, toppled over, and died, Leendertz recalls. I was stunned.

The killer, Leendertz and his group reported inNaturein 2004, was anthrax. It later became clear, however, that the cause was not the usual anthrax bacterium, but an unusual variant ofBacillus cereus, a soil bacterium that is usually benign. But this variant had acquired two circles of DNA, called plasmids, that had turned it into a formidable killer.

Subsequent work showed the bacterium was attacking other Ta forest mammals, too, including monkeys, mongooses, and porcupines. In 2017, the team published evidencegathered from bones, carcasses, and even fliesthat it appeared to be associated with 38% of 279 deaths the team had investigated from 1996 to 2015. The work was a reminder, Leendertz says, that we understand very little about what animals really die of in an environment like this.

Most worrying, theNaturepaper presented simulations showing anthrax could help wipe out the Ta forests chimpanzees within 150 years. And anthrax is not the only disease threatening the chimps, other work by Leendertzs team has shown. On top of all of the deforestation, the poaching they are just getting bashed by these infectious diseases, says primatologist Kimberley Hockings of the University of Exeter.

Some of those deadly diseases come from humans, Leendertz and colleagues reported in 2008 inCurrent Biology. After investigating five respiratory disease outbreaks that had struck Ta chimpanzees between 1999 and 2006, killing at least 15 individuals, the researchers concluded they were linked to two viruses that commonly cause mild disease in humans: human respiratory syncytial virus and human metapneumovirus. Our results suggest that the close approach of humans to apes, which is central to both research and tourism programs, represents a serious threat to wild apes, they wrote.

In Ivory Coast, veterinarian Fabian Leendertz traps bats to test for Ebola virus.

The idea was not new. Jane Goodall, the prominent primatologist, had described a pneumonia outbreak that killed several chimpanzees; researchers believed it was caused by a human-introduced pathogen. But theCurrent Biologystudy, and a similar viral outbreak documented in Tanzania, highlighted the threat of what Goldberg calls reverse zoonoses. Its a world of viruses that are crossing species in every direction, he says. And whenever that happens, it can cause devastating losses. (Goldberg has shown that the most common human cold virus, rhinovirus C, caused a deadly 2013 outbreak among chimpanzees in Uganda.)

The 2008 study also presented a dilemma for primate researchers such as Boesch, who was one of the co-authors. It suggested that even as they studied and worked to protect apes, they might be killing them, too. So, to reduce the risk of future outbreaks, the Ta researchers imposed new restrictions: Incoming staff must quarantine at the camp for 5 days before going into the forest, and everyone must stay at least 7 meters from study animals as well as wear masks while observing. Leendertz, meanwhile, pushed hard for field sites and tourism firms elsewhere to adopt similar measures, co-authoring safety guidelines published in 2015.

Such efforts really opened peoples eyes [that we needed] to be a lot more careful, Hockings says. But, It was a very controversial thing before COVID, Goldberg adds. People were afraid that tourists would be angry if you tried to make them wear a mask, that the apes would be afraid of the masks and attack tourists that foreign governments would get less money from tourism.

Today, Leendertz says helping catalyze such practical, real-world change is among his proudest accomplishments. And he says the experience only underscored the value of long-term, multifaceted studies of wildlife mortality. The threat that infectious diseases pose to chimpanzees was long underestimated and hardly studied, he says. They were neglected for a long time.

DESPITE HIS LOVE of fieldwork, Leendertz is spending less time in the Ta forest these days, visiting just once or twice a year. When my feet are hurting because Im not used to the long distances anymore, and when I get up in the morning from that moldy mattress, I do think that time is over, he says. Still, he says, When I arrive it really is that feeling of coming home.

At the Robert Koch Institute, meanwhile, Leendertzs lab is busy with samples shipped by colleagues in the forest. Located in a brand-new building that also houses one of the worlds newest biosafety level four high-biosecurity labs, the lab uses state-of-the-art technologies to identify and characterize the pathogens found in the samples. Ironically, Leendertz notes, Im back to doing PCRs. Recently, for example, the samples collected from the dead duiker in 2019 were analyzed. The antelope was, as suspected, infected with anthrax.

A research team heads into Kanankru, Ivory Coast, to search for bats, which have been implicated in outbreaks of Ebola and other deadly diseases.

Such molecular sleuthing isnt just about identifying animal killers. Leendertz notes that, when paired with careful field observations, lab findings can yield important insights into protecting human health. In 2017, for instance, some Ta chimpanzees began to cough and display respiratory distress. Lab work showed the cause was monkeypox, a less deadly relative of smallpox that can move from primates to humans. In humans, monkeypox often announces itself through a skin rash, but Leendertzs work suggests coughing is an unusual symptom that health workers working in communities near primate populations should keep in mind.

More recently, Leendertzs team has discovered that leprosyanother disease with the potential to jump to humansaffects wild chimpanzees too. In 2017, Hockings, who studies chimpanzees in Guinea-Bissaus Cantanhez National Park, observed animals with lesions on their faces and hands. She shared her observations with Leendertz, and soon afterward he noticed similar lesions on Woodstock, a Ta chimpanzee. By analyzing fecal samples, the researchers confirmed the lesions were caused by leprosy, a disease never before seen in wild chimpanzees.

The discovery has highlighted how little is known aboutMycobacterium leprae, the bacterium that causes leprosy, says immunologist John Spencer of Colorado State University, Fort Collins. Researchers cant culture the microbe in the laboratory and, although they have found it circulating in armadillos and red squirrels, it hadnt been seen in apes. The chimp find suggests leprosy has other niches that it has adapted to, Spencer saysand adds one more pathogen to the growing roster of diseases that afflict both humans and other animals.

If Leendertz has builthis career on the dual concerns of human and chimpanzee health, then the emergence of SARS-CoV-2 has brought these two issues together with new urgency. The virus now rampaging through human populations is a potential threat to great apes as well, Leendertz and primatologist Tom Gillespie of Emory University warned in a letter published inNaturein March 2020. To reduce the risks, they asked governments to suspend ecotourism and researchers to reduce field research, and many complied.

Since then, gorillas at the San Diego Zoo have tested positive for SARS-CoV-2. They showed only mild symptoms, but that is not very reassuring, Gillespie says, because captive animals tend to be well fed and be less burdened with other infections. Its really hard to say from captive studies what we would see in the wild, he says.

Looking ahead, Leendertz says, The question is how to get back to a more normal situation for primate scientists. One concrete step could be to vaccinate researchers and people living around field sites like Ta, he suggests.

In the meantime, WHO has asked Leendertz to join its investigation into the origins of SARS-CoV-2. That appointment makes sense scientifically and politically, colleagues say. Leendertzs years of patient, intensive focus on understanding death in a single rainforest have given him a valuable perspective on how to investigate pathogens hopping from one species to another, as SARS-CoV-2 is believed to have done. And he represents the Robert Koch Institute, Germanys equivalent of the U.S. Centers for Disease Control and Prevention. I see the WHO mission as about 50% actual science and 50% building bridges with Chinese colleagues, Goldberg says. I think Fabian will do well on both fronts.

But Leendertz also knows from past experience with virus hunts that definitive answers can be hard to come by. In 2014, he led a team that traveled to Meliandou, Guinea, shortly after the start of an Ebola outbreak that ultimately killed some 1000 people. The researchers interviewed villagers, who told them about a hollow tree where the child who had been the first to get sick had played.

When the team visited the tree, they discovered it had burned (whether by accident or intention wasnt clear). On the blackened stump, they found traces of DNA left behind by bats that had apparently roosted in the tree. Had an encounter between the child and a bat sparked the outbreak? It was a plausible scenario, they concluded, but there would likely never be proof.

The chain of events that led to the COVID-19 pandemic is likely to be far more elusive. And the WHO investigation has gotten off to a bumpy start. When the team first tried to visit China earlier this month, officials barred several members from entering because of pandemic restrictions. Leendertz himself could not join the trip because of a family commitment. So, while his colleagues conducted Zoom meetings from the hotel rooms where they were quarantined after arriving in China, Leendertz joined from his home, where it was 2:30 a.m. It was another kind of breakfast TV, just not the episode he enjoys the most.

Read the rest here:
This wildlife vet tracks deadly microbes in the African jungle. Now, he's on the trail of COVID-19 - Science Magazine

Read More...

Police report associated with former LSU player’s 2020 arrest reveals alleged animal abuse – WBRZ

January 31st, 2021 2:51 am

BATON ROUGE Former LSU defensive end, Ray Parker, bonded out jail Thursday (September 10) after being arrested on-campus for battery of a dating partner. Records indicate that Parker's bond amount was $4,500.

According to a police report, he was arrested on LSU campus around 2:30 a.m., Wednesday for allegedly abusing his girlfriend and destroying personal items in her apartment, including an iPhone valued at $1,600.

Arrest records obtained by WBRZ say the two had been dating for about a year, but got into an argument shortly before the altercation, which led to Parker becoming enraged and destroying items around her apartment in addition to pushing her into a dresser, leaving a scrape on her hip.

Police say the woman's injuries and the damage to her apartment and belongings were consistent with her story, so Parker was arrested and booked on charges of battery of a dating partner and criminal damage to property.

Hours after the arrest, LSU head football Coach Ed Orgeron announced that Parker was cut from the team for violating its rules.

Later documents were obtained that shed additional light on the events surrounding Parker's arrest.

A police report dated as "filed" on September 17, 2020 says Parker's former girlfriend accused Parker of beating up their puppy, a pit bull named 'Kash.'

According to the report, she accused Parker of breaking one of the puppy's ribs and legs in addition to threatening her via text message, saying that Kash might be dead by the time she arrived at his apartment.

The girlfriend went on to tell detectives that due to a lack of funds necessary to cover Kash's medical expenses, she turned the puppy over to the LSU School of Veterinary Medicine. Police say they followed up with LSU SVM and found that the young pit bull's leg had to be amputated due to multiple breaks and fractures.

A roommate, according to the police report, also confirmed accusations against Parker in connection with the abuse of the puppy, saying they'd seen Parker kick Kash on multiple occasions.

The roommate said they would often hear the puppy crying and would tell Parker to "chill out."

Parker, 20 years old at the time, was in his second year with the Tigers after redshirting as a true freshman in 2019.

Read this article:
Police report associated with former LSU player's 2020 arrest reveals alleged animal abuse - WBRZ

Read More...

Using Dogs and DNA to Diagnose Diarrhea in Foals The Horse – TheHorse.com

January 31st, 2021 2:51 am

The pictures arent pretty, and the condition is serious. But what truly stinks about foal diarrhea is that in more than half the cases, veterinarians never identify the underlying cause. Yet, according to Nathan Slovis, DVM, Dipl. ACVIM, CHT, director of the McGee Center at Hagyard Equine Medical Institute, in Lexington, Kentucky, that scenario is changing. Developing technologiesand even the use of certified sniffer dogs that can identify Clostridium difficile in humansare making it faster and easier to diagnose whether the culprit is viral, bacterial, protozoal, or something else.

Twenty percent of foals will get some form of infectious diarrhea, Slovis said during his presentation at the 2020 American Association of Equine Practitioners Convention, held virtually. And because so many possible causes exist, veterinarians often find themselves treating symptoms without really knowing what theyre up against. Pathogens such as rotavirus, clostridium, and salmonella can proliferate in neonates with lethal speed. Fortunately, veterinarians can now use real-time polymerase chain reaction (PCR) testing to swiftly identify viruses, bacteria, and protozoa by amplifying fragments of genetic material in blood and fecal samples. Slovis explained that by recording how many cycles it takes to replicate the DNA to a trillion copies, the test also offers a way to quantify infectious loads.

For too long, veterinarians had to rely on what they could see under the microscope or what they could culture in a petri dish. Cultures are often misleading, because potentially harmful bacteria can be found everywhere, even in perfectly healthy horses. Real-time PCR has been a diagnostic game-changer in both human and veterinary medicine, he said.

Routinely treating foals with antibiotics has become controversial, Slovis acknowledged. Antibiotics are ineffective against viruses, and theres growing concern about antibiotic-resistant bacteria, including multidrug resistant strains of salmonella. Genetically engineered bacteria-eating viruses known as bacteriophages hold future promise for treating some of these antibiotic-resistant infections, but more research is needed.

Antibiotics do change the gastrointestinal (GI) tract microbiome, leaving researchers to wonder whether they simply make diarrhea worse. Nonetheless, Slovis generally favors giving sick foals a broad-spectrum antibiotic even when he suspects and confirms rotavirus.

Neonates are different than adults, he said. Bacteremia (bacteria in the bloodstream) occurs in 50% of foal diarrhea cases. Because endotoxins can be deadly, his strategy is to quell any primary or secondary bacterial infections before the foals body is overwhelmed.

Slovis also made a strong pitch for vaccinating pregnant mares against rotavirus and salmonella. A two-dose vaccine for Salmonella typhimurium and agona (conditional license in Kentucky) can be given to mares at nine and 10 months of gestation or to foals of unvaccinated mares during their first month of life. The rotavirus vaccine requires three doses administered at eight, nine, and 10 months of gestation.

Studies show the rotavirus vaccine:

Unfortunately, the currently available rotavirus vaccine only protects from the G3 strain, not the G14 strain thats becoming more common in Kentucky and elsewhere, Slovis said. He hopes a pharmaceutical company might be persuaded to work on a vaccine offering crossover protection between these two equine strains.

Rotavirus hits young foals hard and fast, with a one- to two-day incubation period. The virus invades the GI tract and impacts the production of lactase, which interferes with milk digestion, leading to diarrhea and painful bloating. Using PCR is the best way to diagnose it. Immunoassay tests can also be helpful, he noted, but they must be validated for horses, because human rotavirus antigen tests can provide false negatives in equines. Therapies include providing fluids, ulcer medication, pain relievers, and lactase to support digestion, reduce bloating, and calm the bowels.

Rotavirus shedding can continue for up to 10 days after the babys manure firms up, and the virus can persist in the environment for up to nine months, he cautioned. Bleach is not an effective disinfectant against the virus, whereas alcohol and hydrogen peroxide formulations are, he said.

Bacteria can also plague young foals, with C. difficile, C. perfringens, and Salmonella common culprits. Slovis warned that types A and C of C. perfringens are associated with fatal foal necrotizing enterocolitis (damage and death of cells in the small intestine and colon), so theres no time to waste. By the time a positive culture comes back from the lab, it could be too late. Again, he said PCR is the new gold standard for identifying bacterial loads, possibly providing results within hours.

Lateral flow enzyme immunoassay tests that screen for both antigens and toxins are also useful. Theyre quick and easy, but sometimes produce ambiguous results, such as indicating positive for antigens but negative for toxins.

We used to believe that if we didnt see that a test was positive for a C. difficile toxin, then it wasnt significant, Slovis said. Yet after studying many antigen-positive/toxin-negative foals (all of which had diarrhea, and all which responded to the antibiotic metronidazole), he found PCR tests confirmed more than three-quarters of them did have C. difficile infections.

Yet PCR isnt the only trending diagnostic development. Slovis noted that canines are being trained and certified to sniff out C. diff and other infections. These special technicians, however, are currently in short supply in human medicine.

Hygiene is hugely important in preventing or reducing foal diarrhea outbreaks. If barns are potentially infected, he suggests foaling mares outdoors until all stall surfaces, tack, and equipment have been thoroughly disinfected. Foaling kits should contain only single-use items and equipment that can be sterilized between uses. Slovis also recommended using disposable udder wipes to cleanse the mares udder and hindquarters before her baby nurses. A bath before or after foaling might also be in order. He also emphasized proper handwashing for those handling mares and foals. Soap and towels (preferably touchless), along with hand sanitizer, should be within easy reach and installed where they wont be contaminated, which means not just sitting on the counter next to the sink.

His recommendations also include:

Not every case of foal diarrhea is preventable. But faster, more accurate diagnostics reduce guesswork regarding treatment, leading to better outcomes for babies.

Read this article:
Using Dogs and DNA to Diagnose Diarrhea in Foals The Horse - TheHorse.com

Read More...

Page 218«..1020..217218219220..230240..»


2025 © StemCell Therapy is proudly powered by WordPress
Entries (RSS) Comments (RSS) | Violinesth by Patrick