82 with Everolimus in Patients with Solid Tumors at the ASCO Annual Meeting 2016

On June 5, 2016 Tyrogenex, a privately held biopharmaceutical company, reported that data from its Phase I study of X-82, an oral dual anti-VEGFR/PDGFR tyrosine kinase inhibitor, with everolimus in solid tumors were presented at the American Society of Clinical Oncology (ASCO) (Free ASCO Whitepaper) Annual Meeting 2016 held in Chicago, Illinois (Press release, Tyrogenex, JUN 5, 2016, View Source [SID:1234513093]).

Schedule your 30 min Free 1stOncology Demo!
Discover why more than 1,500 members use 1stOncology™ to excel in:

Early/Late Stage Pipeline Development - Target Scouting - Clinical Biomarkers - Indication Selection & Expansion - BD&L Contacts - Conference Reports - Combinatorial Drug Settings - Companion Diagnostics - Drug Repositioning - First-in-class Analysis - Competitive Analysis - Deals & Licensing

                  Schedule Your 30 min Free Demo!

The data presented were from the Phase I study in which X-82 was combined with full dose everolimus in solid tumors to determine the dose limiting toxicities and recommended Phase II dose for pNET and RCC. Results were presented by Dr. Benjamin R. Tan, MD, Washington University School of Medicine, in a poster titled "Phase I study of X-82, an oral dual anti-VEGFR/PDGFR tyrosine kinase inhibitor, with everolimus in solid tumors."

"The results from this study provide critical information for moving forward with the development of X-82 in solid tumors," said Dr. Tan. "We have also seen favorable responses in renal cell carcinoma (RCC) and prolonged stable diseases (SD) in neuroendocrine tumors (NET) patients, which warrant further investigations in these tumor types. Tyrogenex continued to advances this program with the goal of providing patients suffering from a variety of solid tumors with a safe and effective treatment option."

About the Phase I Study

A 3+3 dose escalation design was utilized to determine the dose limiting toxicities and the recommended Phase II dose of daily oral X-82 plus everolimus at 10 mg PO daily for patients with solid malignancies. Key eligibility criteria included PS 0-1, measurable disease adequate organ function and normal LVEF.

Key conclusions from the study include:

Recommended Phase II dose is X-82 at 300 mg PO daily plus everolimus at 10 mg PO daily
Dose limiting toxicities include Grade 3 fatigue, mucositis and hypophosphatemia
Encouraging responses and prolonged stable disease seen in RCC and NETs
An expansion cohort for RCC and PNET are ongoing at Washington University and Vanderbilt University
About X-82

Tyrogenex’s lead compound is X-82. X-82 inhibits both VEGF and PDGFR. Tyrogenex believes X-82 targets the basic mechanisms of neovascular eye diseases, including angiogenesis, fibrosis and inflammation. X-82 is currently being evaluated for wet Age-Related Macular Degeneration (AMD) and solid tumors.

Foundation Medicine Presents New Data at ASCO 2016 Demonstrating that FoundationOne® May Help Predict Response to Cancer Immunotherapy Across a Variety of Advanced Cancers

On June 5, 2016 Foundation Medicine, Inc. (NASDAQ:FMI) and it collaborators reported data showing that higher tumor mutational burden, as estimated by comprehensive genomic profiling with FoundationOne, successfully predicted a greater likelihood of response and longer response duration to cancer immunotherapies in patients with advanced bladder cancer and metastatic melanoma as well as several other tumor types (Press release, Foundation Medicine, JUN 5, 2016, View Source [SID:1234513005]). The results were presented in two oral sessions, and several poster discussions at the 2016 Annual Meeting of the American Society of Clinical Oncology (ASCO) (Free ASCO Whitepaper) taking place in Chicago.

Schedule your 30 min Free 1stOncology Demo!
Discover why more than 1,500 members use 1stOncology™ to excel in:

Early/Late Stage Pipeline Development - Target Scouting - Clinical Biomarkers - Indication Selection & Expansion - BD&L Contacts - Conference Reports - Combinatorial Drug Settings - Companion Diagnostics - Drug Repositioning - First-in-class Analysis - Competitive Analysis - Deals & Licensing

                  Schedule Your 30 min Free Demo!

"Successful application of cancer immunotherapy in the clinic is one of the most important advances in cancer treatment in decades," said Vincent Miller, M.D., chief medical officer at Foundation Medicine. "These data suggest that tumor mutational burden could serve as an independent predictive biomarker to aid clinicians in identifying patients who are most likely to benefit from cancer immunotherapies that target either the PD-1 or PD-L1 proteins."

Cancer immunotherapy works by helping the immune system mount an effective anti-cancer response, a process that depends in part on the recognition of cancer-specific proteins called neoantigens. Tumor mutational burden has been shown to correlate well with the number of neoantigens, and therefore it may help identify patients most likely to respond to cancer immunotherapies. By combining comprehensive genomic profiling of 315 genes utilizing the FoundationOne assay, with Foundation Medicine’s advanced and proprietary algorithm that filters out normal individual genomic variants, FoundationOne can reliably and accurately measure tumor mutational burden without the need for whole exome sequencing. Foundation Medicine expects to provide a CLIA-certified version of tumor mutational burden on all FoundationOne and FoundationOne Heme reports to physicians in the third quarter 2016.

Overview of Data Presentations

The IMvigor 210 study of TecentriqTM (atezolizumab; anti-PD-L1; Genentech/Roche) in locally advanced or metastatic urothelial carcinoma evaluated three separate biomarkers: PD-L1 protein expression as measured by immunohistochemistry, molecular subtype as measured by gene expression and The Cancer Genome Atlas, and mutational load (often referred to as tumor mutational burden) as measured by FoundationOne. All three biomarkers were shown to be independent predictors of response to Tecentriq.

"PD-L1 Expression, Cancer Genome Atlas (TCGA) Subtype and Mutational Load are Independent Predictors of Response to Atezolizumab (atezo) in Metastatic Urothelial Carcinoma (mUC; Imvigor210)", by Jonathan E. Rosenberg, M.D., Memorial Sloan Kettering Cancer Center [Abstract #104, Clinical Science Symposium, Sunday June 5, 9:57-10:09 AM].
"These results are particularly exciting given the amount of variability inherent to using immunohistochemistry (IHC) to measure biomarkers. There are many different PD-L1 IHC tests, for example, and pathologists often do not see agreement between them," stated Vamsidhar Velcheti, M.D., assistant professor, Solid Tumor Oncology, Taussig Cancer Institute, Cleveland Clinic. "We need a truly quantitative and reproducible approach to predicting response to immunotherapies, and measuring tumor mutational burden using FoundationOne may provide us with that solution."

In a separate melanoma study, higher mutational burden as measured by FoundationOne was associated with a greater likelihood of response and a more durable response to pembrolizumab; anti-PD-1; Merck, nivolumab; anti-PD-1; Bristol Myers Squibb, and Tecentriq, thereby providing oncologists with greater confidence in the potential for clinical benefit from a host of newly approved immunotherapies.

"Hybrid Capture-Based Next Generation Sequencing (HC NGS) in Melanoma Identifies Markers of Response to Anti-PD1/PD-L1", by Douglas Buckner Johnson, M.D., M.S.C.I., Vanderbilt-Ingram Cancer Center [Abstract #105, Clinical Science Symposium, Sunday June 5, 10:21-10:33 AM]
Dr. Miller continued, "The clinical reality is that some patients respond very well to cancer immunotherapies and others do not. As a result, the ability to leverage our molecular information platform to identify the right candidates for these immunotherapies is an important advance for the field of precision medicine. Matching the right therapy with the right patient has the potential to both improve outcomes and increase efficiency in the current care model."

These conclusions were further supported by two additional presentations at ASCO (Free ASCO Whitepaper) 2016 that demonstrated that total mutational burden could also predict response to cancer immunotherapy in lung and colorectal cancers:

"Total Mutational Burden (TMB) in Lung Cancer and Relationship with Response to PD-1/PD-L1 Targeted Therapies", by David R. Spigel, M.D., Sarah Cannon Research Institute [Abstract #9017, Poster Session, Saturday June 4, 8:00-11:30 AM].
"Tumor Mutational Burden as a Potential Biomarker for PD1/PD-L1 Therapy in Colorectal Cancer", by Thomas J. George, M.D., F.A.C.P., University of Florida [Abstract #3587, Poster Session, Saturday June 4, 8:00-11:30 AM].

Daratumumab (DARZALEX®) Combination Therapy Significantly Extended Progression-Free Survival in Previously Treated Patients with Multiple Myeloma

On June 5, 2016 Johnson & Johnson reported that data from the Phase 3 MMY3004 (CASTOR) clinical trial show the immunotherapy daratumumab (DARZALEX) in combination with a standard of care therapy, bortezomib (a proteasome inhibitor [PI]) and dexamethasone (a corticosteroid), demonstrated a 61 percent reduction in the risk of disease progression or death (progression-free survival, or PFS) compared to bortezomib and dexamethasone alone in patients with multiple myeloma who received a median of two prior lines of therapy (Hazard Ratio (HR)=0.39; 95 percent CI (0.28-0.53), p<0.0001) (Press release, Johnson & Johnson, JUN 5, 2016, View Source [SID:1234513094]).1

Schedule your 30 min Free 1stOncology Demo!
Discover why more than 1,500 members use 1stOncology™ to excel in:

Early/Late Stage Pipeline Development - Target Scouting - Clinical Biomarkers - Indication Selection & Expansion - BD&L Contacts - Conference Reports - Combinatorial Drug Settings - Companion Diagnostics - Drug Repositioning - First-in-class Analysis - Competitive Analysis - Deals & Licensing

                  Schedule Your 30 min Free Demo!

According to results Janssen-Cilag International NV announced today, daratumumab also significantly increased the overall response rate (ORR) [83 percent vs. 63 percent, p<0.0001]. The median PFS in the daratumumab arm has not been reached, compared with a median PFS of 7.16 months for patients who received bortezomib and dexamethasone alone.1

These data will be presented in full today at 3:10 – 3:25 p.m. CDT during the "Plenary Session: Including the Science of Oncology Award and Lecture" at the 2016 American Society of Clinical Oncology (ASCO) (Free ASCO Whitepaper) Annual Meeting in Chicago. They have also been selected for inclusion in the ASCO (Free ASCO Whitepaper) Press Programme. In addition, these results will also be featured in an encore, oral presentation at the 21st Annual Congress of the European Hematology Association (EHA) (Free EHA Whitepaper) on Sunday 12 June at 12:00 – 12:15 p.m. CEST (Abstract #LB2236).

"We saw clinically meaningful improvements in progression-free survival and overall response rates with daratumumab when combined with standard of care," said Antonio Palumbo, M.D., Myeloma Unit Chief, Department of Oncology, Division of Haematology, University of Torino, Italy. "These compelling Phase 3 results demonstrate that a regimen built on daratumumab deepens clinical responses and help to underscore its potential for multiple myeloma patients who have been previously treated."

In addition to meeting the primary endpoint of improved PFS at a median follow-up of 7.4 months and significantly increasing the ORR compared to bortezomib and dexamethasone alone, daratumumab doubled rates of complete response (CR) or better [19 percent vs. 9 percent, p=0.0012], including doubling rates of very good partial response (VGPR) [59 percent vs. 29 percent, p<0.0001]. The median PFS has not been reached, compared with a median PFS of 7.16 months for patients who received bortezomib and dexamethasone alone. The treatment benefit of the daratumumab combination regimen was maintained across clinically relevant subgroups.1

"At Janssen we are committed to redefining the impact cancer has on patients, through delivering innovative research and solutions. We’re therefore extremely encouraged by the remarkable interim results of this study. The findings provide an important insight into the effect daratumumab can have in combination with established regimens, and illustrate the promise of this immunotherapy in earlier lines of treatment," said Jane Griffiths, Company Group Chairman, Janssen Europe, Middle East and Africa. "We’re dedicated to exploring the full treatment value of daratumumab for multiple myeloma patients and look forward to the difference we can make with data like these."

Overall, the safety of the daratumumab combination therapy was consistent with the known safety profile of daratumumab monotherapy (D) and bortezomib plus dexamethasone (Vd), respectively. The most common (>25 percent) adverse events (AEs) [DVd/Vd] were thrombocytopenia (59 percent/44 percent), peripheral sensory neuropathy (47 percent/38 percent), diarrhea (32 percent/22 percent) and anaemia (26 percent/31 percent). Most common grade 3 or 4 AEs (>10 percent) were thrombocytopenia (45 percent/33 percent), anaemia (14 percent/16 percent) and neutropenia (13 percent/4 percent). The rate of Grade 3/4 infections/infestations was 21 percent in the DVd group and 19 percent in the Vd group. The most common Grade 3/4 infections/infestations treatment-emergent AEs, or TEAEs (≥5 percent) was pneumonia (8 percent/10 percent). The number of patients with Grade 3 or 4 bleeding events (3 patients in DVd group, 2 patients in Vd group) was low in both treatment groups. Few (7 percent/9 percent) patients discontinued therapy due to a TEAE.1

About the MMY3004 (CASTOR) Trial

The Phase 3, multinational, open-label, randomised, multicentre, active-controlled MMY3004 study has included 498 patients with multiple myeloma who received a median of two prior lines of therapy. Sixty-six percent of patients received prior treatment with bortezomib; 76 percent received prior treatment with an immunomodulatory agent; and 48 percent received prior treatment with a PI and immunomodulatory agent. Thirty-three percent of patients were refractory to an immunomodulatory agent, and 32 percent were refractory to their last line of prior therapy. Patients were randomised to receive either daratumumab combined with subcutaneous bortezomib and dexamethasone (n=251) or bortezomib and dexamethasone alone (n=247). Participants were treated with daratumumab until disease progression, unacceptable toxicity, or if they had other reasons to discontinue the study.1

On March 30, 2016, the MMY3004 (CASTOR) trial was unblinded after meeting its primary endpoint of improved PFS in a pre-planned interim analysis (HR = 0.39, p<0.0001). Based on the recommendation of an Independent Data Monitoring Committee (IDMC), patients in the standard of care treatment arm were offered the option to receive daratumumab following confirmed disease progression.2

Janssen will initiate discussions with regulatory authorities about the potential for a regulatory submission for this indication based on the results of this study. A comprehensive clinical study report is being prepared for submission to global health authorities.

Additional Combination Data

The Phase 3 MMY3003 (POLLUX) study, comparing daratumumab in combination with lenalidomide and dexamethasone versus lenalidomide and dexamethasone alone in patients with multiple myeloma who received at least one prior line of therapy, was also unblinded in May 2016. Based on the results at the pre-planned interim analysis conducted by an IDMC, the study met its primary endpoint of improved PFS.3 The POLLUX data have been selected for inclusion in the Presidential Symposium at EHA (Free EHA Whitepaper) on Friday 10 June 2016 at 4:47 p.m. CEST (Abstract #LB2238).

About Daratumumab

Daratumumab is a first-in-class biologic targeting CD38, a surface protein that is highly expressed across multiple myeloma cells, regardless of disease stage.4-6 Daratumumab induces rapid tumour cell death through apoptosis (programmed cell death)7,8 and multiple immune-mediated mechanisms of action, including complement-dependent cytotoxicity (CDC), antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP).7,9,10 Daratumumab has also demonstrated immunomodulatory effects that contribute to tumour cell death via a decrease in immune suppressive cells including T-regs, B-regs and myeloid-derived suppressor cells.7,11 Five Phase 3 clinical studies with daratumumab in relapsed and frontline settings are currently ongoing. Additional studies are ongoing or planned to assess its potential in other malignant and pre-malignant diseases on which CD38 is expressed. For more information, please see www.clinicaltrials.gov.

In May 2016, daratumumab was approved by the European Commission (EC) for monotherapy of adult patients with relapsed and refractory multiple myeloma (MM), whose prior therapy included a proteasome inhibitor (PI) and an immunomodulatory agent and who have demonstrated disease progression on the last therapy. Daratumumab was approved under an accelerated assessment, a process reserved for medicinal products expected to be of major public health interest, particularly from the point of view of therapeutic innovation.12

In August 2012, Janssen Biotech, Inc. and Genmab A/S entered a worldwide agreement, which granted Janssen an exclusive license to develop, manufacture and commercialise daratumumab.

About Multiple Myeloma

Multiple myeloma is an incurable blood cancer that occurs when malignant plasma cells grow uncontrollably in the bone marrow.13,14 Refractory cancer occurs when a patient’s disease is resistant to treatment or in the case of multiple myeloma, patients progress within 60 days of their last therapy.15,16 Relapsed cancer means the disease has returned after a period of initial, partial or complete remission.17 Accounting for approximately one percent of all cancers and 15 percent to 20 percent of haematologic malignancies worldwide,18 multiple myeloma is designated as an orphan disease in both Europe and the US. Globally, it is estimated that 124,225 people were diagnosed, and 87,084 died from the disease in 2015.19,20 While some patients with multiple myeloma have no symptoms at all, most patients are diagnosed due to symptoms which can include bone fracture or pain, low red blood counts, fatigue, calcium elevation, kidney problems or infections.14 Patients who relapse after treatment with standard therapies (including PIs or immunomodulatory agents) typically have poor prognoses and few remaining options.21

New Data from Phase 2 Study Evaluating KEYTRUDA® (pembrolizumab) for the Treatment of Cancers Deficient in DNA Mismatch Repair Show Durable Responses Across a Range of Cancers

On June 5, 2016 Merck (NYSE: MRK), known as MSD outside the United States and Canada, reported updated findings from a study evaluating KEYTRUDA (pembrolizumab), the company’s anti-PD-1 therapy, in patients with advanced cancers characterized as deficient for DNA mismatch repair (MMR) (Press release, Merck & Co, JUN 5, 2016, View Source [SID:1234513008]). Results showed that among previously treated patients with MMR-deficient tumors, there was an overall response rate (ORR) of 53 percent (n=16/30) (95% CI, 36-70) in patients with a range of advanced non-colorectal solid tumors and an ORR of 57 percent (n=16/28) (95% CI, 39-73) in patients with advanced colorectal cancer; in contrast, no responses were observed in patients with advanced colorectal cancer whose tumors were characterized as MMR-proficient (n=0/25). The findings, from a phase 2 study led by researchers from Johns Hopkins Kimmel Cancer Center in collaboration with Merck, were presented at the 52nd Annual Meeting of the American Society of Clinical Oncology (ASCO) (Free ASCO Whitepaper) in Chicago (Abstracts #3003, #103).

Schedule your 30 min Free 1stOncology Demo!
Discover why more than 1,500 members use 1stOncology™ to excel in:

Early/Late Stage Pipeline Development - Target Scouting - Clinical Biomarkers - Indication Selection & Expansion - BD&L Contacts - Conference Reports - Combinatorial Drug Settings - Companion Diagnostics - Drug Repositioning - First-in-class Analysis - Competitive Analysis - Deals & Licensing

                  Schedule Your 30 min Free Demo!

"We continue to be encouraged by the findings of this ongoing study evaluating KEYTRUDA in patients with mismatch repair deficient tumors," said Dr. Roger Dansey, senior vice president and therapeutic area head, oncology late-stage development, Merck Research Laboratories. "Characterization of biomarkers with the potential to predict clinical outcomes for patients receiving immune therapy for cancer is an important element of our clinical program for KEYTRUDA."

Merck is conducting a phase 2 registration study (KEYNOTE-164) to evaluate the efficacy and safety of KEYTRUDA monotherapy in patients with previously treated, locally advanced unresectable or metastatic (Stage IV) MMR-deficient or microsatellite instability-high (MSI-H) colorectal cancer and a phase 3 study (KEYNOTE-177) in a treatment-naïve patient population. An additional phase 2 clinical trial (KEYNOTE-158) is evaluating patients with advanced tumors classified as MSI-H, excluding colorectal carcinoma.

The KEYTRUDA (pembrolizumab) clinical development program includes more than 30 tumor types in more than 270 clinical trials, including more than 100 trials that combine KEYTRUDA with other cancer treatments.

Updated Findings from Non-Colorectal Cancer Cohort (Abstract #3003) and Colorectal Cancer Cohort (Abstract #103)

The phase 2 study evaluated the clinical activity of KEYTRUDA monotherapy (10 mg/kg every two weeks) in patients with previously treated, progressive metastatic disease with or without MMR-deficiency. Three groups were evaluated: MMR-deficient non-colorectal cancers (n=30), MMR-deficient colorectal cancer (n=28), and MMR-proficient colorectal cancer (n=25). MMR status was assessed locally using a standard immunohistochemistry (IHC) or polymerase chain reaction (PCR)-based method for detection of microsatellite instability. The key endpoints of the study were ORR, duration of response (DOR), progression-free survival (PFS) measured by RECIST v1.1, and overall survival (OS).

Abstract #3003 described findings from a cohort of patients with MMR-deficient non-colorectal cancers (including ampullary/biliary, endometrial, gastric, pancreatic, prostate, sarcoma, and small bowel) who received KEYTRUDA. Results demonstrated an ORR of 53 percent (n=16/30) (95% CI, 36-70) – 30 percent were complete responses (n=9/30) and 23 percent were partial responses (n=7/30). Additionally, 17 percent of patients had stable disease (n=5/30), and the disease control rate was 70 percent (n=21/30) (95% CI, 52-83). Median PFS and OS were not reached at the time of analysis; median duration of follow-up was 10 months.

Abstract #103 described findings from two cohorts of patients with colorectal cancer (MMR-deficient and MMR-proficient) who received KEYTRUDA. Among those patients with MMR-deficient tumors, an ORR of 57 percent was observed (n=16/28) (95% CI, 39-73) – 11 percent were complete responses (n=3/28) and 46 percent were partial responses (n=13/28). Additionally, 32 percent of patients had stable disease (n=9/28) and the disease control rate was 89 percent (n=25/28) (95% CI, 73-96). The median PFS and OS were not reached; median duration of follow-up was 9.3 months. Among patients with MMR-proficient tumors, no responses were observed (n=0/25) and the disease control rate was 16 percent (n=4/25) (95% CI, 6-35); 16 percent had stable disease (n=4/25). Additionally, in patients with MMR-proficient tumors, the median PFS was 2.3 months and the median OS was 5.98 months; median duration of follow-up was 6 months.

The safety profile of KEYTRUDA (pembrolizumab) was consistent with that observed in previously reported studies. For patients in the MMR-deficient non-colorectal cancers cohort, Grade 3-4 treatment-related adverse events included diarrhea/colitis (n=3) and pancreatitis (n=1). For patients in the colorectal cancer cohorts (MMR-deficient and MMR-proficient), Grade 3-4 treatment-related adverse events included diarrhea/colitis (n=1), pancreatitis (n=2), rash/pruritus (n=1), anemia (n=1), leukopenia (n=1), and thrombocytopenia (n=1).

Initial findings from this study evaluating KEYTRUDA (41 patients) were presented at the 2015 ASCO (Free ASCO Whitepaper) Annual Meeting and simultaneously published online in the New England Journal of Medicine (Le et al. New Eng. J. Med. 372, 26, 2509). On November 2, 2015, Merck announced that the U.S. Food and Drug Administration (FDA) granted Breakthrough Therapy Designation to KEYTRUDA for the treatment of patients with MSI-H metastatic colorectal cancer.

About DNA Mismatch Repair and Microsatellite Instability

Analysis of tumor DNA for microsatellite instability can be used to identify tumors with defective DNA mismatch repair (MMR) systems. DNA MMR is a process that in normal cells permits the recognition and repair of genetic mismatches generated during DNA replication. A defective MMR system results in the persistence of DNA mismatches that may then be incorporated into the genetic code as mutations. The average tumor has dozens of mutations; however, tumors with DNA MMR-deficiency may harbor thousands, especially in regions of repetitive DNA known as microsatellites. Tumors that are found to have mutations in select microsatellite sequences, called microsatellite instability (MSI), are considered DNA MMR-deficient. These tumors are referred to as being "MSI-high." Overall, DNA MMR-deficiency is present in approximately 15-20 percent of tumors in Stage II disease, 10 percent in Stage III disease and approximately 5 percent or less in Stage IV disease. In colorectal cancers, MMR-deficiency is seen in approximately 15-20 percent of non-hereditary colorectal cancers and in most hereditary colorectal cancers associated with Lynch Syndrome.

About KEYTRUDA (pembrolizumab) Injection 100 mg

KEYTRUDA is a humanized monoclonal antibody that works by increasing the ability of the body’s immune system to help detect and fight tumor cells. KEYTRUDA 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.

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

KEYTRUDA is also indicated for the treatment of patients with metastatic non-small cell lung cancer (NSCLC) whose tumors express PD-L1 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. This indication is approved under accelerated approval based on tumor response rate and durability of response. An improvement in survival or disease-related symptoms has not yet been established. Continued approval for this indication may be contingent upon verification and description of clinical benefit in the confirmatory trials.

KEYTRUDA is administered at a dose of 2 mg/kg as an intravenous infusion over 30 minutes every three weeks for the approved indications.

Selected Important Safety Information for KEYTRUDA (pembrolizumab)

Immune-mediated pneumonitis, including fatal cases, occurred in patients receiving KEYTRUDA. Pneumonitis occurred in 32 (2.0%) of 1567 patients with melanoma, including Grade 1 (0.8%), 2 (0.8%), and 3 (0.4%) pneumonitis. Pneumonitis occurred in 19 (3.5%) of 550 patients with NSCLC, including Grade 2 (1.1%), 3 (1.3%), 4 (0.4%), or 5 (0.2%) pneumonitis and more frequently in patients with a history of asthma/chronic obstructive pulmonary disease (5.4%) or prior thoracic radiation (6.0%). Monitor patients for signs and symptoms of pneumonitis. Evaluate suspected pneumonitis with radiographic imaging. Administer corticosteroids for Grade 2 or greater pneumonitis. Withhold KEYTRUDA for Grade 2; permanently discontinue KEYTRUDA for Grade 3 or 4 or recurrent Grade 2 pneumonitis.

Immune-mediated colitis occurred in 31 (2%) of 1567 patients with melanoma, including Grade 2 (0.5%), 3 (1.1%), and 4 (0.1%) colitis. Immune-mediated colitis occurred in 4 (0.7%) of 550 patients with NSCLC, including Grade 2 (0.2%) or 3 (0.4%) colitis. Monitor patients for signs and symptoms of colitis. Administer corticosteroids for Grade 2 or greater colitis. Withhold KEYTRUDA for Grade 2 or 3; permanently discontinue KEYTRUDA for Grade 4 colitis.

Immune-mediated hepatitis occurred in patients receiving KEYTRUDA. Hepatitis occurred in 16 (1%) of 1567 patients with melanoma, including Grade 2 (0.1%), 3 (0.7%), and 4 (0.1%) hepatitis. Monitor patients for changes in liver function. Administer corticosteroids for Grade 2 or greater hepatitis and, based on severity of liver enzyme elevations, withhold or discontinue KEYTRUDA.

Hypophysitis occurred in 13 (0.8%) of 1567 patients with melanoma, including Grade 2 (0.3%), 3 (0.3%), and 4 (0.1%) hypophysitis. Hypophysitis occurred in 1 (0.2 %) of 550 patients with NSCLC, which was Grade 3 in severity. Monitor patients for signs and symptoms of hypophysitis (including hypopituitarism and adrenal insufficiency). Administer corticosteroids and hormone replacement as clinically indicated. Withhold KEYTRUDA (pembrolizumab) for Grade 2; withhold or discontinue for Grade 3 or 4 hypophysitis.

Hyperthyroidism occurred in 51 (3.3%) of 1567 patients with melanoma, including Grade 2 (0.6%) and 3 (0.1%) hyperthyroidism. Hypothyroidism occurred in 127 (8.1%) of 1567 patients with melanoma, including Grade 3 (0.1%) hypothyroidism. Hyperthyroidism occurred in 10 (1.8%) of 550 patients with NSCLC, including Grade 2 (0.7%) or 3 (0.3%) hyperthyroidism. Hypothyroidism occurred in 38 (6.9%) of 550 patients with NSCLC, including Grade 2 (5.5%) or 3 (0.2%) hypothyroidism. Thyroid disorders can occur at any time during treatment. Monitor patients for changes in thyroid function (at the start of treatment, periodically during treatment, and as indicated based on clinical evaluation) and for clinical signs and symptoms of thyroid disorders. Administer replacement hormones for hypothyroidism and manage hyperthyroidism with thionamides and beta-blockers as appropriate. Withhold or discontinue KEYTRUDA (pembrolizumab) for Grade 3 or 4 hyperthyroidism.

Type 1 diabetes mellitus, including diabetic ketoacidosis, occurred in 3 (0.1%) of 2117 patients. Monitor patients for hyperglycemia or other signs and symptoms of diabetes. Administer insulin for type 1 diabetes, and withhold KEYTRUDA and administer anti hyperglycemics in patients with severe hyperglycemia.

Immune-mediated nephritis occurred in patients receiving KEYTRUDA. Nephritis occurred in 7 (0.4%) of 1567 patients with melanoma including, Grade 2 (0.2%), 3 (0.2%), and 4 (0.1%) nephritis. Monitor patients for changes in renal function. Administer corticosteroids for Grade 2 or greater nephritis. Withhold KEYTRUDA for Grade 2; permanently discontinue KEYTRUDA for Grade 3 or 4 nephritis.

Other clinically important immune-mediated adverse reactions can occur. For suspected immune-mediated adverse reactions, ensure adequate evaluation to confirm etiology or exclude other causes. Based on the severity of the adverse reaction, withhold KEYTRUDA and administer corticosteroids. Upon improvement to Grade 1 or less, initiate corticosteroid taper and continue to taper over at least 1 month. Based on limited data from clinical studies in patients whose immune-related adverse reactions could not be controlled with corticosteroid use, administration of other systemic immunosuppressants can be considered. Resume KEYTRUDA when the adverse reaction remains at Grade 1 or less following corticosteroid taper. Permanently discontinue KEYTRUDA (pembrolizumab) for any Grade 3 immune-mediated adverse reaction that recurs and for any life-threatening immune-mediated adverse reaction.

The following clinically significant, immune-mediated adverse reactions occurred in less than 1% (unless otherwise indicated) of 1567 patients with melanoma: arthritis (1.6%), exfoliative dermatitis, bullous pemphigoid, uveitis, myositis, Guillain-Barré syndrome, myasthenia gravis, vasculitis, pancreatitis, hemolytic anemia, and partial seizures arising in a patient with inflammatory foci in brain parenchyma. The following clinically significant, immune-mediated adverse reactions occurred in less than 1% of 550 patients with NSCLC: rash, vasculitis, hemolytic anemia, serum sickness, and myasthenia gravis.

Severe and life-threatening infusion-related reactions have been reported in 3 (0.1%) of 2117 patients. Monitor patients for signs and symptoms of infusion related reactions including rigors, chills, wheezing, pruritus, flushing, rash, hypotension, hypoxemia, and fever. For Grade 3 or 4 reactions, stop infusion and permanently discontinue KEYTRUDA (pembrolizumab).

Based on its mechanism of action, KEYTRUDA can cause fetal harm when administered to a pregnant woman. If used during pregnancy, or if the patient becomes pregnant during treatment, apprise the patient of the potential hazard to a fetus. Advise females of reproductive potential to use highly effective contraception during treatment and for 4 months after the last dose of KEYTRUDA.

In Trial 6, KEYTRUDA was discontinued due to adverse reactions in 9% of 555 patients with advanced melanoma; adverse reactions leading to 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%). Adverse reactions leading to interruption of KEYTRUDA occurred in 21% of patients; the most common (≥1%) was diarrhea (2.5%). The most common adverse reactions with KEYTRUDA vs ipilimumab were fatigue (28% vs 28%), diarrhea (26% with KEYTRUDA), rash (24% vs 23%), and nausea (21% with KEYTRUDA). Corresponding incidence rates are listed for ipilimumab only for those adverse reactions that occurred at the same or lower rate than with KEYTRUDA.

In Trial 2, KEYTRUDA was discontinued due to adverse reactions in 12% of 357 patients with advanced melanoma; the most common (≥1%) were general physical health deterioration (1%), asthenia (1%), dyspnea (1%), pneumonitis (1%), and generalized edema (1%). Adverse reactions leading to interruption of KEYTRUDA occurred in 14% of patients; the most common (≥1%) were dyspnea (1%), diarrhea (1%), and maculo-papular rash (1%). The most common adverse reactions with KEYTRUDA vs chemotherapy were fatigue (43% with KEYTRUDA), pruritus (28% vs 8%), rash (24% vs 8%), constipation (22% vs 20%), nausea (22% with KEYTRUDA), diarrhea (20% vs 20%), and decreased appetite (20% with KEYTRUDA). Corresponding incidence rates are listed for chemotherapy only for those adverse reactions that occurred at the same or lower rate than with KEYTRUDA (pembrolizumab).

KEYTRUDA was discontinued due to adverse reactions in 14% of 550 patients with NSCLC. Serious adverse reactions occurred in 38% of patients. The most frequent serious adverse reactions reported at least 2% of patients were pleural effusion, pneumonia, dyspnea, pulmonary embolism, and pneumonitis. The most common adverse reactions (reported in at least 20% of patients) were fatigue (44%), cough (29%), decreased appetite (25%), and dyspnea (23%).

No formal pharmacokinetic drug interaction studies have been conducted with KEYTRUDA.

It is not known whether KEYTRUDA is excreted in human milk. Because many drugs are excreted in human milk, instruct women to discontinue nursing during treatment with KEYTRUDA and for 4 months after the final dose.

Safety and effectiveness of KEYTRUDA have not been established in pediatric patients.

Our Focus on Cancer

Our goal is to translate breakthrough science into innovative oncology medicines to help people with cancer worldwide. At Merck Oncology, helping people fight cancer is our passion and supporting accessibility to our cancer medicines is our commitment. Our focus is on pursuing research in immuno-oncology and we are accelerating every step in the journey – from lab to clinic – to potentially bring new hope to people with cancer.

As part of our focus on cancer, Merck is committed to exploring the potential of immuno-oncology, with one of the fastest-growing development programs in the industry. We are currently executing an expansive research program that includes more than 270 clinical trials evaluating our anti-PD-1 therapy across more than 30 tumor types. We also continue to strengthen our immuno-oncology portfolio through strategic acquisitions and prioritizing the development of several promising immunotherapeutic candidates with the potential to improve the treatment of advanced cancers.

For more information about our oncology clinical trials, visit www.merck.com/clinicaltrials.

AbbVie Presents Encouraging Phase 1 Data for Investigational Medicine ABT-414 as Monotherapy in Patients with an Aggressive Brain Cancer at the 2016 American Society of Clinical Oncology Annual Meeting

On June 5, 2016 AbbVie (NYSE: ABBV), a global biopharmaceutical company, reported safety and preliminary efficacy data from a Phase 1 study of ABT-414 – an investigational antibody drug conjugate (ADC) for epidermal growth factor receptor (EGFR) amplified, recurrent glioblastoma (GBM) – showed no dose-limiting toxicities and frequent, reversible ocular toxicities (Press release, AbbVie, JUN 5, 2016, View Source [SID:1234513029]). Additionally, an estimated 30 percent (n=44) of patients treated with ABT-414 as monotherapy were progression free at six months [95% CI=17, 44] (secondary endpoint). These results, from an expansion cohort of one arm (Arm C) of a three-arm open-label study, were presented today at the 52nd Annual Meeting of the American Society of Clinical Oncology (ASCO) (Free ASCO Whitepaper) in Chicago.1 Amplified EGFR is the most common genetic mutation associated with malignant GBM, an aggressive brain cancer.2

Schedule your 30 min Free 1stOncology Demo!
Discover why more than 1,500 members use 1stOncology™ to excel in:

Early/Late Stage Pipeline Development - Target Scouting - Clinical Biomarkers - Indication Selection & Expansion - BD&L Contacts - Conference Reports - Combinatorial Drug Settings - Companion Diagnostics - Drug Repositioning - First-in-class Analysis - Competitive Analysis - Deals & Licensing

                  Schedule Your 30 min Free Demo!

As of January 7, 2016, the most common serious adverse event (>1 patient) (n=48) was seizure (8%). Additionally, Best Response Assessment in Neuro-Oncology (RANO) Criteria, an assessment of tumor response used in GBM, identified two partial responses, 18 patients with stable disease, and 24 with progressive disease for a total of 44 patients with complete data.1

"With standard of care therapy,3 patients with GBM, the most common and most aggressive form of brain cancer,4 have a median survival of 15 months after diagnosis and two-year survival is 30%.5 There remains an urgent unmet need for new treatment options for this devastating brain cancer," said Martin van den Bent, M.D., Ph.D., head, Neuro-Oncology Unit, Erasmus MC Cancer Institute, the Netherlands, and lead investigator of the study. "These data are important as they demonstrate the potential of ABT-414 and underscore the need for further investigation in glioblastoma."

Additional Safety Findings
Grade 3/4 treatment emergent adverse events (TEAEs) (>1 patient) were keratitis (15%), corneal epithelial microcysts (8%), hemiparesis (6%), hyperglycemia (6%), muscular weakness (6%), seizure (6%), blurred vision (4%) and ulcerative keratitis (4%).The most common TEAEs (?25% patients) in this study arm were blurred vision (60%), headache (29%), photophobia (29%), dry eye (27%), eye pain (27%), and fatigue (27%). The most common serious adverse event (>1 patient) (n=48) was seizure (8%).1

"These results suggest that ABT-414 may have important activity for certain groups of patients with glioblastoma and support the continuation of the ongoing randomized studies," said Gary Gordon, M.D., vice president, oncology clinical development, AbbVie. "AbbVie is committed to continuing to invest in technologies and approaches, including antibody drug conjugates like ABT-414, with the goal of delivering a remarkable impact on cancer treatment."

Based on these results, together with previously presented data from this study, AbbVie advanced ABT-414 to a randomized Phase 2 clinical trial in patients with EGFR-amplified GBM.1

About this Study
The Phase 1, open-label trial was designed to evaluate the safety, pharmacokinetics and maximum tolerated dose of ABT-414. Three study arms evaluated ABT-414 with radiotherapy and temozolomide (TMZ) in patients with newly diagnosed glioblastoma (GBM) (Arm A), with TMZ in patients with newly diagnosed glioblastoma who have just completed radiation and TMZ or recurrent GBM (Arm B) or as monotherapy in patients with recurrent GBM (Arm C).1

Eligible patients in Arm C were adults with a Karnofsky Performance Status (KPS) score ?70, EGFR amplification (confirmed centrally), recurrent GBM, normal end-organ function and no prior treatment with bevacizumab. Forty-eight EGFR-amplified recurrent GBM patients were treated in this arm. The median age was 59 years (range, 35-80). Most patients had prior therapies: 40 percent had one, 48 percent had two, and 10 percent had three or more prior therapies.1

About ABT-414
ABT-414 is an investigational monoclonal antibody drug conjugate (ADC) targeting EGFR (epidermal growth factor receptor) developed by AbbVie researchers with components in-licensed from Life Science Pharmaceuticals, Inc. and Seattle Genetics.1 It is being evaluated for the treatment of patients with EGFR-amplified glioblastoma, an aggressive malignant primary brain tumor.1,4 In 2014, the FDA and the European Medicines Agency granted orphan drug designation for the treatment of glioblastoma and glioma, respectively.6,7 ABT-414 is an investigational compound and its efficacy and safety have not been established by the FDA or any other health authority.

About Glioblastoma
Glioblastoma is the most common and most aggressive type of malignant primary brain tumor.4 Mutations in EGFR are the most common genetic abnormality associated with glioblastoma, with a frequency of about 50 percent.2 Prior to diagnosis, patients may experience headache, vision problems, nausea/vomiting, personality changes and seizures.4 For adults with more aggressive glioblastoma, treated with standard therapy, median survival is about 15 months.5 Treatment for glioblastoma remains challenging.4 Standard treatment is surgical resection, radiotherapy and concomitant adjunctive chemotherapy.5