On October 6, 2015 Innate Pharma SA (the "Company" – Euronext Paris: FR0010331421 – IPH) reported the opening of the Phase I/II trial of IPH2201, a first-in-class NKG2A checkpoint inhibitor, tested in combination with ibrutinib in patients with relapsed or refractory Chronic Lymphocytic Leukemia ("CLL") (Press release, Innate Pharma, OCT 6, 2015, View Source [SID:1234507648]). This trial, which will include up to 45 patients, is multicentric and will be performed in the United States. Schedule your 30 min Free 1stOncology Demo! Pierre Dodion, Chief Medical Officer of Innate Pharma, said: "HLA-E is expressed by CLL cells of virtually all patients. IPH2201 is a new checkpoint inhibitor targeting both T and NK cells and preventing their inhibition by HLA-E on tumor cells. In addition, ibrutinib has been demonstrated to create a favorable pro-inflammatory environment; this could result in a synergistic effect with the immunomodulating action of IPH2201". He added: "The Ohio State University Comprehensive Cancer Center is a leading center in developing new therapies to cure Chronic Lymphocytic Leukemia. It is a great opportunity for the development of IPH2201 to work with them".
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!
Pr. John Byrd, Director, and Dr Farrukh Awan, Principal Coordinating Investigator, both expert leaders in the field of CLL at the Division of Hematology, Department of Internal Medicine, Ohio State University, said: "Ibrutinib represents a breakthrough medicine that was approved for the treatment of relapsed and del(17(p13.1) CLL in 2014. However, although ibrutinib induces a high response rate in patients with CLL, responses are rarely complete. Ultimately the disease progresses in a number of patients. Achieving complete responses would be of great interest to potentially prolong remission, and maybe eventually improve survival rate. Targeting the immune system in several novel ways is the rationale to combine IPH2201 and ibrutinib in this trial".
This trial is part of a global co-development and commercialization agreement with AstraZeneca for IPH2201. Within this frame, Innate Pharma expects to have four trials opened by the end of 2015. In addition to the CLL trial, two Phase I/II studies are currently ongoing, testing IPH2201 as a single agent respectively in squamous cell carcinoma of the Head and Neck and in Ovarian cancer . The fourth trial, testing IPH2201 in combination cetuximab in patients with Head and Neck cancer, will start in the coming months.
The co-development plan also includes Phase II combination clinical trials with IPH2201 and durvalumab (MEDI4736), a PD-L1 immune checkpoint inhibitor, in solid tumors, which will be performed by AstraZeneca/MedImmune.
About study IPH2201-202:
This Phase Ib/IIa study is a multicenter open label trial of the combination of IPH2201 and ibrutinib in patients with relapsed or refractory Chronic Lymphocytic Leukemia. Its primary objective is to evaluate the anti-leukemic activity of the combination and the primary endpoint for efficacy is complete response rate. The secondary objectives are to assess the safety of the combination of IPH2201 and ibrutinib. The trial will be performed in the United States under the coordination of leading investigators at the Ohio State University.
36 to 45 patients are planned to be enrolled. The trial is conducted in two parts:
In the first part of the study, 12 to 24 patients will receive a combination of ibrutinib at the approved dosage and IPH2201; 4 dose levels of IPH2201 up to 10 mg/kg will be explored. Based on previous experience with IPH2201, these dosages are expected to induce saturation of the NKG2A receptor.
In the second part of the study, IPH2201 at the dose selected in the dose-escalating part will be assessed in combination with ibrutinib during 26 cycles in up to 24 patients.
The rationale of this trial is based on the observation that HLA-E is expressed in virtually all patients with CLL, at higher levels compared to normal B cells (Veuillen, Aurran-Schleinitz et al. 2012). IPH2201 is a NGK2A checkpoint inhibitor that blocks the HLA-E driven inhibition of NK and CD8+ cells. By binding to NGK2A, IPH2201 restores the capability of those cells to destroy tumor cells. Furthermore, ibrutinib has been demonstrated to create a favorable pro-inflammatory environment; this could result in a synergistic effect with the immunotherapeutic action of IPH2201. Thus, treatment with IPH2201 in combination with ibrutinib may improve the quality of response above and beyond that achieved with ibrutinib alone and achieve complete responses; a higher rate of complete response should lead to improved overall survival.
In a Phase I dose-escalation safety trial, IPH2201 was found to be safe and well-tolerated.
About Chronic Lymphocytic Leukemia (CLL):
CLL results from progressive accumulation of morphologically mature B lymphocytes in the blood, bone marrow and lymphatic tissues. In Western countries, CLL is the most common form of leukemia, accounting for about 25% of all leukemias. Incidence increases with age and the median age of diagnosis is 70 for males and 74 for females. It is estimated that 15,720 new cases will occur in 2014 in the US, causing 4,600 deaths (Siegel, Ma et al., 2014).
Ibrutinib, a first in class kinase inhibitor of BCR signaling, has been approved in 2014 for the treatment of patients with CLL who have received at least one prior therapy. Its approval was based on the safety and efficacy results of several trials which have shown mainly partial responses. The indication for ibrutinib was subsequently extended to include CLL with 17p deletion, irrespectively of the line of therapy.
About IPH2201:
IPH2201 is a first-in-class immune checkpoint inhibitor targeting NKG2A receptors expressed on tumor infiltrating cytotoxic CD8 T lymphocytes and NK cells.
NKG2A is an inhibitory receptor binding HLA-E. By expressing HLA-E, cancer cells can protect themselves from killing by NKG2A+ immune cells. HLA-E is frequently up-regulated on cancer cells of many solid tumors or hematological malignancies. IPH2201, a humanized IgG4, blocks the binding of NKG2A to HLA-E allowing activation of NK and cytotoxic T cell responses.
Hence, IPH2201 may re-establish a broad anti-tumor response mediated by NK and T cells. IPH2201 may also enhance the cytotoxic potential of other therapeutic antibodies. IPH2201 is partnered with AstraZeneca and MedImmune, the Company’s global biologics research and development arm, through a co-development and commercialization agreement. The initial
development plan includes: Phase II combination clinical trials with durvalumab (MEDI4736) in solid tumors; multiple Phase II trials planned by Innate Pharma to study IPH2201 both as monotherapy and in combination with currently approved treatments across a range of cancers; and the development of associated biomarkers. As previously announced, under the terms of this agreement, Innate Pharma is eligible to cash payments of up to $1.275 billion as well as double digit royalties on sales. In addition to the initial payment of $250 million to Innate Pharma, AstraZeneca will pay a further $100 million prior to initiation of Phase III development, as well as additional regulatory and sales-related milestones of up to $925 million. AstraZeneca will book all sales and will pay Innate Pharma double-digit royalties on net sales. The arrangement includes the right for Innate Pharma to co-promote in Europe for a 50% profit share in the territory.
Author: [email protected]
GTx Announces Enrollment of First Patient in Phase 2 Clinical Trial of Enobosarm in Triple Negative Breast Cancer
On October 6, 2015 GTx, Inc. (Nasdaq: GTXI) reported the enrollment of the first patient into its Phase 2 clinical trial of enobosarm (GTx-024) to treat women with advanced, androgen receptor positive (AR+), triple negative breast cancer (TNBC) (Press release, GTx, OCT 6, 2015, View Source [SID:1234507646]). Enobosarm is the Company’s lead product candidate and is also being evaluated in a separate Phase 2 clinical trial to treat estrogen receptor positive (ER+), AR+ breast cancer, which the Company recently announced had also enrolled its first patient. Schedule your 30 min Free 1stOncology Demo! "Most women with triple negative breast cancer have extremely limited treatment options and poor prognoses," said Robert J. Wills, Ph.D., Executive Chairman of GTx. "Based on our preclinical research and positive data from patient-derived and cell line-derived xenografts of TNBC, we are hopeful that enobosarm, by targeting the androgen receptor, may offer another treatment option to women with this disease."
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 open-label, multi-center, multinational Phase 2 clinical trial (NCT02368691) will evaluate the efficacy and safety of orally administered enobosarm in up to 55 women with advanced, AR+ TNBC. Patients will receive 18 mg of enobosarm once daily for up to 12 months. The initial stage will be assessed among the first 21 evaluable patients. If at least 2 of 21 patients achieve clinical benefit at week 16, then the trial will proceed to the second stage of enrollment of up to a total of 41 evaluable patients. Clinical benefit is defined as a complete response, partial response, or stable disease, as measured by Response Evaluation Criteria in Solid Tumors (RECIST) at 16 weeks. The trial, which is being conducted under the leadership of Dr. Hope Rugo from the University of California at San Francisco, will include investigators from more than 40 clinical trial sites in the U.S. and abroad.
About enobosarm
Enobosarm, a selective androgen receptor modulator (SARM), has been evaluated in multiple completed or ongoing clinical trials enrolling over 1,500 subjects at doses ranging from 0.1 mg to 100 mg. At all evaluated dose levels, enobosarm was observed to be generally safe and well tolerated.
Most recently, enobosarm 9 mg has been tested in a Phase 2, proof of concept clinical trial of 22 postmenopausal women with ER+ metastatic breast cancer who have previously responded to endocrine therapy. Seventeen of the 22 patients were confirmed to be AR+. Six of these 17 patients demonstrated clinical benefit at six months. Seven patients in total (one patient with indeterminate AR status) achieved clinical benefit at six months. The results also demonstrated that, after a median duration on study of 81 days, 41 percent of all patients (9/22) achieved clinical benefit as best response and also had increased serum PSA levels which may be an indicator of AR activity. Enobosarm was well tolerated. The most common adverse events reported were pain, fatigue, nausea, hot flash/night sweats, and arthralgia.
About Triple Negative Breast Cancer
Breast cancer is the most commonly diagnosed cancer in women and one in eight women will develop invasive breast cancer in their lifetime. In 2012, 1.7 million women were diagnosed with breast cancer, and there were 6.3 million women alive who had been diagnosed with breast cancer in the previous five years. Clinical assessment of breast cancer includes routine characterization of receptor status including the presence or absence of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) in the tumor tissue. Receptor status is used to assess metastatic potential as well as to guide treatment decisions. Although the majority of breast cancers are considered hormone receptor positive, expressing ER and/or PR, 15–20 percent of women diagnosed with breast cancer will have triple negative breast cancer (TNBC), which is characterized by a lack of expression of ER, PR, and HER2. TNBC occurs more frequently in younger patients (< 50 years of age) and generally shows a more aggressive behavior. For those patients with advanced TNBC, standard palliative treatment options are limited to cytotoxic chemotherapy. However, even after initial response to chemotherapy, the duration of the response may be short, and there is a higher likelihood of visceral metastases, rapidly progressing disease, and inferior survival compared to hormone positive breast cancer. Therefore, research is focused on identifying therapeutic targets in TNBC.
Studies have demonstrated that up to 50 percent of TNBC will express the androgen receptor. Both preclinical and patient-derived and cell line-derived AR+ TNBC xenografts support the clinical approach of targeting the androgen receptor with enobosarm.
Exelixis Announces Positive Overall Survival Results from Phase 3 Pivotal Trial of Cobimetinib in Combination with Vemurafenib in Patients with BRAF V600 Mutation-Positive Advanced Melanoma
On October 6, 2015 Exelixis, Inc. (NASDAQ:EXEL) reported positive overall survival (OS) results from coBRIM, the phase 3 pivotal trial evaluating cobimetinib, a specific MEK inhibitor discovered by Exelixis, in combination with vemurafenib in previously untreated patients with unresectable locally advanced or metastatic melanoma carrying a BRAF V600 mutation (Press release, Exelixis, OCT 6, 2015, View Source [SID:1234507644]). Schedule your 30 min Free 1stOncology Demo! Exelixis’ collaborator Genentech, a member of the Roche Group, informed the company that coBRIM met its secondary endpoint of demonstrating a statistically significant and clinically meaningful increase in overall survival for patients receiving the combination of cobimetinib and vemurafenib, as compared to vemurafenib monotherapy. Ongoing study monitoring did not identify any new safety signals. Long-term safety data are expected later this year. These data will be the subject of a presentation at an upcoming medical meeting.
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 positive effect of the combination of cobimetinib and vemurafenib on overall survival is a major step forward for patients with advanced BRAF V600 mutation-positive melanoma in search of new treatment options," said Michael M. Morrissey, Ph.D., president and chief executive officer of Exelixis. "We continue to work with our partners at Genentech in preparation for the potential U.S. approval and launch of cobimetinib to deliver this important potential treatment option to patients, physicians and caregivers in the United States as quickly as possible."
Exelixis announced the first regulatory approval of cobimetinib in Switzerland in August 2015. U.S. and EU regulatory applications sponsored by Genentech and Roche, respectively, are currently under review. In the United States, the Prescription Drug User Fee Act action date is November 11, 2015. In the EU, Roche anticipates a regulatory decision by the end of this year following a positive opinion from the European Committee for Medicinal Products for Human Use announced in late September.
About the coBRIM Study
The coBRIM trial is an international, randomized, double-blind, placebo-controlled Phase III study evaluating the safety and efficacy of 60 mg once daily of cobimetinib in combination with 960 mg twice daily of vemurafenib, compared to 960 mg twice daily of vemurafenib alone. In the study, 495 patients with BRAF V600 mutation-positive unresectable locally advanced or metastatic melanoma (detected by the cobas 4800 BRAF Mutation Test) and previously untreated for advanced disease, were randomized to receive vemurafenib every day on a 28-day cycle plus either cobimetinib or placebo for days 1-21. Treatment was continued until disease progression, unacceptable toxicity or withdrawal of consent. Investigator-assessed PFS is the primary endpoint. Secondary endpoints include PFS by independent review committee, overall response rate, overall survival, duration of response and other safety, pharmacokinetic and quality of life measures.
About the Cobimetinib Development Collaboration
Exelixis discovered cobimetinib internally and advanced the compound to investigational new drug (IND) status. In late 2006, Exelixis entered into a worldwide co-development agreement with Genentech, under which Exelixis received initial upfront and milestone payments for signing the agreement and submitting the IND. Exelixis was responsible for development of cobimetinib through the end of phase 1, at which point Genentech exercised its option to further develop the compound.
If cobimetinib is approved in the United States, Exelixis is entitled to an initial equal share of U.S. profits and losses, which will decrease as sales increase, and will share in U.S. marketing and commercialization costs. In November 2013, Exelixis exercised its option to co-promote cobimetinib in the United States and, under the terms of the agreement, the company is prepared to field up to 25 percent of the U.S. sales force.
About the Combination of Cobimetinib and Vemurafenib
Cobimetinib is a selective inhibitor that blocks the activity of MEK, a protein kinase that is part of a key pathway (the RAS-RAF-MEK-ERK pathway) that promotes cell division and survival. This pathway is frequently activated in human cancers including melanoma, where mutation of one of its components (BRAF) causes abnormal activation in about 50 percent of tumors. About 50 percent of patients with BRAF mutation positive melanoma experience a tumor response when treated with a BRAF inhibitor, however development of resistance and subsequent tumor progression limits treatment benefit. Clinical and preclinical analyses indicated that reactivation of the MEK-ERK pathway may underlie development of resistance to BRAF inhibitors in many progressing tumors, and that co-treatment with a BRAF and MEK inhibitor delays the emergence of resistance in the preclinical setting, providing the rationale for testing the combination of vemurafenib and cobimetinib in clinical trials. In addition to the combination with vemurafenib in melanoma, cobimetinib is also being investigated in combination with several investigational medicines, including an immunotherapy, in several tumor types, including non-small cell lung cancer, colorectal cancer, triple-negative breast cancer and melanoma.
About Melanoma and its BRAF V600 Mutation-Positive Form
Melanoma is the less common, but more serious category of skin cancer that starts in the skin’s pigment producing cells known as melanocytes. According to the American Cancer Society, approximately five percent of skin cancer diagnoses are melanoma, but melanoma accounts for a large majority of skin cancer deaths. In recent years, there have been significant advances in treatment for metastatic melanoma and people with the disease have more options. However, it continues to be a serious health issue with a high unmet need and a steadily increasing incidence over the past 30 years. It is projected that approximately half of all melanomas, and eight percent of solid tumors, contain a mutation of the BRAF protein. BRAF is a key component of the RAS-RAF-MEK-ERK pathway involved in normal cell growth and survival. However, mutations that keep the BRAF protein in an active state may cause excessive signaling in the pathway, leading to uncontrolled cell growth and survival. The BRAF V600 mutation-positive form of melanoma is associated with high-risk characteristics of the disease, including early onset, the absence of chronic skin damage, and decreased survival.
FDA approves expanded indication for medical device to treat a form of brain cancer
On October 5, 2015 The U.S. Food and Drug Administration reported they approved an expanded indication for the Optune device to treat patients with newly-diagnosed glioblastoma multiforme (GBM), an aggressive form of brain cancer (Press release, , OCT 5, 2015, View Source [SID:1234507650]). It is given along with the chemotherapy drug temozolomide (TMZ) following standard treatments that include surgery, chemotherapy, and radiation therapy.
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!
Nearly 23,000 Americans will be diagnosed with brain or other nervous system cancers in 2015, according to the National Cancer Institute, and more than 15,000 will die from them. GBM accounts for about 15 percent of all brain tumors, and typically occurs in adults ages 45 to 70. Patients survive less than 15 months on average following diagnosis, because the tumor tends to be highly resistant to standard treatments.
"Patients newly diagnosed with this challenging and aggressive form of brain cancer now have another treatment option available," said William Maisel, M.D., M.P.H., acting director of the Office of Device Evaluation in the FDA’s Center for Devices and Radiological Health. "While the treatment is not a cure, it can increase survival by several months."
In the clinical study used to support the expanded indication, patients treated with the device and TMZ lived on average three months longer than those treated with the drug alone.
Optune was initially approved in 2011 to treat patients with GBM that recurred or progressed after chemotherapy. With this expanded indication, Optune can be used as part of a standard treatment for GBM before the disease progresses.
For newly diagnosed GBM, Optune is not intended to be used as a substitute for standard treatments, but rather as an adjunct therapy, and should not be used without a physician’s supervision.
When using Optune, a health care professional places electrodes on the surface of the patient’s scalp to deliver low-intensity, alternating electrical fields called "tumor treatment fields" (TTFields). The unique shape and special characteristics of rapidly dividing tumor cells make them susceptible to damage when exposed to TTFields, which could halt tumor growth.
The device is portable and can be powered with batteries or plugged into an electrical outlet. Patients can use the device at home or work, allowing them to continue their normal daily activities.
The FDA based its approval of the expanded indication of the Optune device on results from a clinical trial involving 695 patients newly diagnosed with GBM that compared those who used Optune with TMZ to those receiving TMZ alone. Patients who used the device along with TMZ lived, on average, about seven months with no disease progression compared to four months for those who had the drug alone. The Optune plus TMZ group survived for an average of 19.4 months after diagnosis compared to 16.6 months for those who were treated with only TMZ.
Optune for newly diagnosed GBM was reviewed under the FDA’s priority review program, which provides for an expedited review of certain devices that treat life-threatening conditions.
The most common side effect experienced with Optune was skin irritation. Clinical trial participants also experienced a slightly higher incidence of neurological side effects, including convulsions and headaches, compared to subjects receiving TMZ alone.
Patients should not use the Optune system if they have an active implanted medical device or a skull defect, have an underlying skin condition involving the scalp or have a known sensitivity to conductive hydrogels, such as those used on electrocardiogram stickers.
The Optune System is made by Novocure Inc. of Portsmouth, New Hampshire.
The FDA, an agency within the U.S. Department of Health and Human Services, protects the public health by assuring the safety, effectiveness, and security of human and veterinary drugs, vaccines and other biological products for human use, and medical devices. The agency also is responsible for the safety and security of our nation’s food supply, cosmetics, dietary supplements, products that give off electronic radiation, and for regulating tobacco products.
Myriad to Present Seven Studies at the ASHG Annual Meeting
On October 5, 2015 Myriad Genetics, Inc. (NASDAQ:MYGN), a leader in molecular diagnostics and personalized medicine, reported that there will be seven poster presentations featured at American Society for Human Genetics (ASHG) meeting being held Oct. 6 to 10, 2015 in Baltimore, Md (Press release, Myriad Genetics, OCT 5, 2015, View Source [SID:1234507643]).
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!
"At Myriad, we are passionately committed to providing the most accurate hereditary cancer tests through extensively validated laboratory and variant interpretation processes. We have spent almost two decades of research to provide the most sophisticated and comprehensive variant classification program," said Rick Wenstrup, M.D., chief medical officer, Myriad Genetics. "We look forward to presenting these seven new studies at ASH (Free ASH Whitepaper)G as we continue to advance the state-of-the-art in variant classification with innovative new approaches."
A list of the Myriad presentations at ASH (Free ASH Whitepaper)G (#ASHG15) follows:
Poster Presentations
Title: RNA functional studies for the classification of germline variants of uncertain significance that may impair splicing.
Date: Wednesday, Oct. 7, 2015: 5:00 to 6:00 p.m. ET.
Location: Poster 2639W.
Title: No evidence for a difference in the severity of cancer history for carriers of missense versus truncating pathogenic variants in ATM.
Date: Thursday, Oct. 8, 2015: 12:00 to 1:00 p.m. ET.
Location: Poster 2750T.
Title: Detection of large rearrangements in a pan-cancer gene panel using next generation sequencing.
Date: Thursday, Oct. 8, 2015: 12:00 to 1:00 p.m. ET.
Location: Poster 2018T.
Title: Comparison of a literature search algorithm and curated publication database with the literature content of other locus specific databases.
Date: Thursday, Oct. 8, 2015: 12:00 to 1:00 p.m. ET.
Location: Poster 1739T.
Title: Analytical validation of a saliva collection and DNA extraction protocol increases accessibility to hereditary pan-cancer panel testing.
Date: Thursday, Oct. 8, 2015: 12:00 to 1:00 p.m. ET.
Location: Poster 1985T.
Title: Identification of retrotransposon insertion mutations in hereditary cancer.
Date: Thursday, Oct. 8, 2015: 12:00 to 1:00 p.m. ET.
Location: Poster 2592T.
Title: Does this patient need to be tested for Lynch Syndrome? Assessing the reliability of family history for ascertainment.
Date: Friday, Oct. 9, 2015: 11:45 a.m. to 12:45 p.m. ET.
Location: Poster 2082F.
For more information about these presentations, including a complete list of abstracts and posters, please visit the ASH (Free ASH Whitepaper)G website at www.ashg.org/2015meeting/pages/online-planner.shtml.