Bristol-Myers Squibb and Halozyme Enter Global Collaboration and License Agreement for ENHANZE Technology

On September 14, 2017 Bristol-Myers Squibb Company (NYSE:BMY) and Halozyme Therapeutics, Inc. (NASDAQ:HALO) reported a global collaboration and license agreement to develop subcutaneously administered Bristol-Myers Squibb immuno-oncology medicines using Halozyme’s ENHANZE drug-delivery technology (Press release, Bristol-Myers Squibb, SEP 14, 2017, View Source [SID1234530558]).

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 are excited to partner with Halozyme to pursue potential new approaches to how our medicines are delivered to patients," said Murdo Gordon, chief commercial officer, Bristol-Myers Squibb. "Through our work with Halozyme, we hope to improve the patient treatment experience by developing flexible and convenient treatment delivery options."

The Halozyme ENHANZE technology is based on a proprietary recombinant human hyaluronidase enzyme (rHuPH20) that temporarily degrades hyaluronan — a glycosaminoglycan or chain of natural sugars in the body — to aid in the dispersion and absorption of other injected therapeutic drugs. This technology may allow for more rapid delivery of large volume injectable medications, such as medications that are currently delivered intravenously, through subcutaneous delivery.

"Bristol-Myers Squibb has one of the industry’s most advanced and extensive immuno-oncology portfolios with a clear commitment to patient-centered innovation," said Dr. Helen Torley, president and chief executive officer of Halozyme. "Through this collaboration we are excited to explore the potential for ENHANZE to expand the number of cancer patients who may receive their therapies as a rapidly administered subcutaneous injection."

Under the terms of the agreement, Halozyme will receive an initial $105 million for access to the ENHANZE technology. Bristol-Myers Squibb has designated multiple immuno-oncology targets including programmed death 1 (PD-1) and has an option to select additional targets within five years from the effective date. The collaboration may extend to a maximum of 11 targets. Halozyme has the potential to earn milestone payments of up to $160 million for each of the nominated collaboration targets and additional milestone payments for combination products, subject to achievement of specified development, regulatory and sales-based milestones. In addition, Bristol-Myers Squibb will pay Halozyme royalties on sales of products using the ENHANZE technology developed under the collaboration.

The agreement is subject to customary anti-trust clearance by the U.S. Justice Department and Federal Trade Commission pursuant to the Hart-Scott-Rodino Act.

For Bristol-Myers Squibb, the transaction is expected to be dilutive to Non-GAAP earnings per share (EPS) in 2017 and 2018 by approximately $0.01, and by approximately $0.05 in 2019.

About ENHANZE Technology

Halozyme’s proprietary ENHANZE drug-delivery technology is based on its patented recombinant human hyaluronidase enzyme (rHuPH20). rHuPH20 has been shown to remove traditional limitations on the volume of biologics that can be delivered subcutaneously (just under the skin). By using rHuPH20, some biologics and compounds that are administered intravenously may instead be delivered subcutaneously. ENHANZE may also benefit subcutaneous biologics by reducing the need for multiple injections. This delivery has been shown in studies to reduce health care practitioner time required for administration and shorten time for drug administration.

10-Q – Quarterly report [Sections 13 or 15(d)]

Oncbiomune has filed a 10-Q – Quarterly report [Sections 13 or 15(d)] with the U.S. Securities and Exchange Commission (Filing, 10-Q, OncBioMune Pharmaceuticals, 2017, SEP 14, 2017, View Source [SID1234522112]).

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!

Halozyme Licenses New Enhanze Target For $30 Million Upfront Payment, Future Milestones And Royalties

On September 14, 2017 Halozyme Therapeutics, Inc. (NASDAQ: HALO) reported it licensed its ENHANZE drug-delivery technology to Roche for exclusive development of an undisclosed therapeutic target (Press release, Halozyme, SEPT 14, 2017, View Source [SID1234527667]).

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!

Under terms of the agreement, Halozyme will receive an initial $30 million with the potential to earn additional payments of up to $160 million subject to achievement of specified development, regulatory and sales-based milestones. Halozyme will also receive tiered, mid-single digit royalties on sales of commercialized products.

"We are pleased to broaden our longstanding collaboration with Roche to include selection of a new target," said Dr. Helen Torley, president and CEO. "With each new licensing agreement, we see the potential for our global partners to advance their innovative therapies, reducing the treatment burden for patients, caregivers and payers through shorter administration times or a less frequent dosing regimen."

The Halozyme/Roche relationship dates back to the original global collaboration and licensing agreement for the ENHANZE technology signed in 2006. From this agreement, Roche developed two subcutaneous formulations of cancer drugs for markets worldwide. More recently, the relationship expanded to include the study of Halozyme’s investigational oncology drug, PEGPH20, with atezolizumab as part of a clinical collaboration announced in 2016.

The Halozyme ENHANZE technology is based on a proprietary recombinant human hyaluronidase enzyme (rHuPH20) that temporarily degrades hyaluronan — a glycosaminoglycan or chain of natural sugars in the body — to aid in the dispersion and absorption of other injected therapeutic drugs. For Halozyme partners, this technology may allow for more rapid delivery of injectable medications through subcutaneous delivery.

About ENHANZE Technology
Halozyme’s proprietary ENHANZE drug-delivery technology is based on its patented recombinant human hyaluronidase enzyme (rHuPH20). rHuPH20 has been shown to remove traditional limitations on the volume of biologics that can be delivered subcutaneously (just under the skin). By using rHuPH20, some biologics and compounds that are administered intravenously may instead be delivered subcutaneously. ENHANZE may also benefit subcutaneous biologics by reducing the need for multiple injections. This delivery has been shown in studies to reduce health care practitioner time required for administration and shorten time for drug administration.

Pfizer and Astellas Announce Positive Top-Line Results from Phase 3 PROSPER Trial of XTANDI (enzalutamide) in Patients with Non-Metastatic Castration-Resistant Prostate Cancer

On September 14, 2017 Pfizer Inc. (NYSE:PFE) and Astellas Pharma Inc. (TSE: 4503, President and CEO: Yoshihiko Hatanaka, "Astellas") reported that the Phase 3 PROSPER trial evaluating XTANDI (enzalutamide) plus androgen deprivation therapy (ADT) versus ADT alone in patients with non-metastatic (M0) Castration-Resistant Prostate Cancer (CRPC) met its primary endpoint of improved metastasis-free survival (MFS) (Press release, Pfizer, SEP 14, 2017, View Source [SID1234520514]). The preliminary safety analysis of the PROSPER trial appears consistent with the safety profile of XTANDI in previous clinical trials.

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!

"Many prostate cancer patients who initiate androgen deprivation therapy will experience disease progression illustrated by a rising PSA level, and currently, there are no FDA-approved treatment options for patients with non-metastatic CRPC until they develop confirmed radiographic metastatic disease," said Neal Shore, M.D., director, CPI, Carolina Urologic Research Center.

Based on the results of PROSPER, the companies intend to discuss the data with global health authorities to potentially support expanding the label for XTANDI to cover all patients with CRPC.

"We are delighted with the significant results seen in the PROSPER study, showing that XTANDI plus ADT delayed clinically detectable metastases compared to ADT alone in patients with non-metastatic CRPC whose only sign of underlying disease was a rapidly rising prostate-specific antigen (PSA) level. We look forward to discussing the data with regulatory authorities," said Mace Rothenberg, M.D., chief development officer, Oncology, Pfizer Global Product Development. "XTANDI is already established as a standard of care for men with metastatic CRPC based on the results of prior studies, such as AFFIRM and PREVAIL, which demonstrated that XTANDI delayed disease progression and improved overall survival in men with clinically detectable metastatic disease."

"We want to thank the patients, family members and clinicians who participated in the PROSPER trial and helped advance the scientific understanding of the potential role for XTANDI in this prevalent disease," said Steven Benner, M.D., senior vice president and global therapeutic area head, oncology development, Astellas. "We look forward to further analyzing the detailed efficacy and safety results from PROSPER, and submitting them for presentation at an upcoming major medical meeting."

As part of Pfizer and Astellas’ ongoing commitment to the clinical development of enzalutamide in areas of greatest unmet need, the companies initiated the PROSPER trial to evaluate the potential benefits of XTANDI in men with non-metastatic CRPC, an earlier stage of prostate cancer where there are currently no FDA-approved treatment options. On June 9, 2017, the companies announced an amendment to the PROSPER protocol, which accelerated the clinical trial completion date by two years.

XTANDI is currently approved for the treatment of metastatic CRPC based on clinical data from previous studies that showed a statistically significant overall survival benefit for XTANDI versus placebo in the metastatic CRPC setting. XTANDI has been prescribed to more than 185,000 patients globally since its first approval in 2012.

About PROSPER

The Phase 3 randomized, double-blind, placebo-controlled, multi-national trial enrolled approximately 1,400 patients with non-metastatic castration-resistant prostate cancer (CRPC) at sites in the United States, Canada, Europe, South America and the Asia Pacific region. PROSPER enrolled patients with prostate cancer that had progressed, based on a rising prostate-specific antigen (PSA) level despite androgen deprivation therapy (ADT), but who had no symptoms with no prior or present evidence of metastatic disease. The primary objective of the trial was metastasis-free survival (MFS). MFS is a measure of the amount of time that passes until a cancer can be radiographically detected as having metastasized, or spread, to other parts of the body. The trial evaluated enzalutamide at a dose of 160 mg taken orally once daily plus ADT, versus placebo plus ADT. For more information on the PROSPER trial go to www.clinicaltrials.gov.

XTANDI has not yet been evaluated by the FDA for the treatment of patients with non-metastatic CRPC.

About Non-Metastatic Castration-Resistant Prostate Cancer

According to the American Cancer Society, more than 161,000 men are estimated to be diagnosed with prostate cancer in 2017.[i] Castration-resistant prostate cancer (CRPC) refers to the subset of men whose prostate cancer progresses despite androgen deprivation therapy.[ii] Non-metastatic CRPC means there is no clinically detectable evidence of the cancer spreading to other parts of the body (metastases), and there is a rising prostate-specific antigen (PSA) level.[iii] Many men with non-metastatic CRPC will go on to develop metastatic CRPC.[iv]

About XTANDI (enzalutamide) capsules

XTANDI (enzalutamide) is an androgen receptor inhibitor that blocks multiple steps in the androgen receptor signaling pathway within the tumor cell. In preclinical studies, enzalutamide has been shown to competitively inhibit androgen binding to androgen receptors, and inhibit androgen receptor nuclear translocation and interaction with DNA. The clinical significance of this mechanism of action (MOA) is unknown.

XTANDI is approved by the U.S. Food and Drug Administration for the treatment of patients with metastatic castration-resistant prostate cancer (mCRPC). Additional ongoing studies, such as the ARCHES trial in metastatic hormone-sensitive prostate cancer and the EMBARK trial in non-metastatic hormone-sensitive prostate cancer, are continuing to evaluate the potential of enzalutamide to help patients in need.

Important Safety Information

Contraindications

XTANDI is not indicated for women. XTANDI can cause fetal harm and potential loss of pregnancy.

Warnings and Precautions

Seizure occurred in 0.5% of patients receiving XTANDI in clinical studies. In a study of patients with predisposing factors, seizures were reported in 2.2% of patients. See section 5.1 of the Prescribing Information for the list of predisposing factors. It is unknown whether anti-epileptic medications will prevent seizures with XTANDI. Permanently discontinue XTANDI in patients who develop a seizure during treatment.

Posterior Reversible Encephalopathy Syndrome (PRES) In post approval use, there have been reports of PRES in patients receiving XTANDI. PRES is a neurological disorder which can present with rapidly evolving symptoms including seizure, headache, lethargy, confusion, blindness, and other visual and neurological disturbances, with or without associated hypertension. A diagnosis of PRES requires confirmation by brain imaging, preferably MRI. Discontinue XTANDI in patients who develop PRES.

Adverse Reactions

The most common adverse reactions (≥ 10%) that occurred more commonly (≥ 2% over placebo) in the XTANDI patients from the two placebo-controlled clinical trials were asthenia/fatigue, back pain, decreased appetite, constipation, arthralgia, diarrhea, hot flush, upper respiratory tract infection, peripheral edema, dyspnea, musculoskeletal pain, weight decreased, headache, hypertension, and dizziness/vertigo. In the bicalutamide-controlled study of chemotherapy-naïve patients, the most common adverse reactions (≥ 10%) reported in XTANDI patients were asthenia/fatigue, back pain, musculoskeletal pain, hot flush, hypertension, nausea, constipation, upper respiratory tract infection, diarrhea, and weight loss.

In the placebo-controlled study of patients taking XTANDI who previously received docetaxel, Grade 3 and higher adverse reactions were reported among 47% of XTANDI patients and 53% of placebo patients. Discontinuations due to adverse events were reported for 16% of XTANDI patients and 18% of placebo patients. In the placebo-controlled study of chemotherapy-naïve patients, Grade 3-4 adverse reactions were reported in 44% of XTANDI patients and 37% of placebo patients. Discontinuations due to adverse events were reported for 6% of both study groups. In the bicalutamide-controlled study of chemotherapy-naïve patients, Grade 3-4 adverse reactions were reported in 38.8% of XTANDI patients and 37.6% of bicalutamide patients. Discontinuations due to adverse events were reported for 7.6% of XTANDI patients and 6.3% of bicalutamide patients.

Lab Abnormalities: In the two placebo-controlled trials, Grade 1-4 neutropenia occurred in 15% of XTANDI patients (1% Grade 3-4) and 6% of placebo patients (0.5% Grade 3-4). Grade 1-4 thrombocytopenia occurred in 6% of XTANDI patients (0.3% Grade 3-4) and 5% of placebo patients (0.5% Grade 3-4). Grade 1-4 elevations in ALT occurred in 10% of XTANDI patients (0.2% Grade 3-4) and 16% of placebo patients (0.2% Grade 3-4). Grade 1-4 elevations in bilirubin occurred in 3% of XTANDI patients (0.1% Grade 3-4) and 2% of placebo patients (no Grade 3-4).

Infections: In the study of patients taking XTANDI who previously received docetaxel, 1% of XTANDI patients compared to 0.3% of placebo patients died from infections or sepsis. In the study of chemotherapy-naïve patients, 1 patient in each treatment group (0.1%) had an infection resulting in death.

Falls (including fall-related injuries) occurred in 9% of XTANDI patients and 4% of placebo patients in the two placebo-controlled trials. Falls were not associated with loss of consciousness or seizure. Fall-related injuries were more severe in XTANDI patients, and included non-pathologic fractures, joint injuries, and hematomas.

Hypertension occurred in 11% of XTANDI patients and 4% of placebo patients in the two placebo-controlled trials. No patients experienced hypertensive crisis. Medical history of hypertension was balanced between arms. Hypertension led to study discontinuation in < 1% of patients in each arm.

Drug Interactions

Effect of Other Drugs on XTANDI Avoid strong CYP2C8 inhibitors, as they can increase the plasma exposure to XTANDI. If co-administration is necessary, reduce the dose of XTANDI.

Avoid strong CYP3A4 inducers as they can decrease the plasma exposure to XTANDI. If co-administration is necessary, increase the dose of XTANDI.

Effect of XTANDI on Other Drugs Avoid CYP3A4, CYP2C9, and CYP2C19 substrates with a narrow therapeutic index, as XTANDI may decrease the plasma exposures of these drugs. If XTANDI is co-administered with warfarin (CYP2C9 substrate), conduct additional INR monitoring.

Please see Full Prescribing Information for additional safety information.

SELLAS’ Galinpepimut-S Induces Specific, Robust and Durable Immune Responses in Patients With High-Risk Multiple Myeloma – Correlated With Clinical Benefit

On September 14, 2017 SELLAS Life Sciences Group Ltd. (SELLAS), a privately-held, oncology-focused, clinical stage biopharmaceutical company, reported that its WT1-targeting immuno-oncology (IO) treatment, galinpepimut-S, led to mounting of specific, potent and durable immune responses (IRs) in multiple myeloma (MM) patients (Press release, Sellas Life Sciences, SEP 14, 2017, View Source;Correlated-With-Clinical-Benefit/default.aspx [SID1234520526]).

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!

Galinpepimut-S, is SELLAS’ lead product candidate and is currently expected to enter a pivotal, Phase 3 clinical trial in patients with AML and is also in various development phases in multiple myeloma and ovarian cancer, and additional indications are expected as a monotherapy or in combination with other immuno-oncology agents. The data reported in this release pertain to its ongoing Phase 2 study evaluating galinpepimut-S as a treatment in MM.

In the ongoing Phase 2 study, both the rate (frequency) and potency of the durable IRs in MM patients were correlated with clinical benefit accorded by galinpepimut-S, hereby defined as achievement of cCR/VGPR in patients who completed per protocol immunizations (12 doses). These novel data were presented yesterday, September 13, 2017 , at the Annual Meeting of the Society of Hematologic Oncology (SOHO) in Houston, Texas , USA ( soho2017.com ) at a poster session and the relevant abstract (#MM-252) was published in the September 2017 supplemental issue of the journal Clinical Lymphoma, Myeloma & Leukemia . These IR results considerably expand on previous clinical observations of notable anti-myeloma activity of this innovative IO agent after upfront induction, melphalan conditioning and successful autologous stem cell transplantation (ASCT).

SELLAS’ Phase 2 MM study has enrolled a total of 20 patients who are being monitored long-term. A current median progression-free survival (PFS) of 23.6 months post-ASCT was previously reported in a group of 18 patients, all of whom had evidence of at least minimal residual disease (MRD+) post-ASCT, with 15/18 also having high-risk cytogenetics at baseline. This group represents a population at extremely high risk for progression and poor long-term outcomes, even in the face of post-ASCT maintenance therapy with immunomodulatory drugs (IMiD’s).

Rates of IRs (CD4 and/or CD8) toward any of galinpepimut-S’s individual WT1 peptides (two heteroclitic and four native), as well as a pool of 113 partially overlapping 15-mers spanning the entire length of WT1 (‘all pool’ reactivity) were high at the time of completion of per protocol immunizations, ranging from 72-91%. The rates of immunization against the cognate native epitopes (122A and WT1A) of the two heteroclitic peptides (122A1 and WT1A1) within galinpepimut-S were also high, thus demonstrating the principle of heteroclitic peptide antigenicity. Multivalent IRs (i.e., IRs against more than one WT1 peptide) were detected in up to 66% of patients. IR against WT1 ‘all pool’ peptides denoted multifunctional cross-epitope T-cell reactivity – akin to epitope spreading- and is a hallmark of an effective, cytotoxicity-inducing vaccine. Both the frequency and potency of CD4 IRs against the four native WT1 peptides targeted by galinpepimut-S, as well as ‘all pool’ WT1 antigens, showed a strong positive correlation with indices of clinical benefit (rate of CR/VGPR) at the time of completion of per protocol immunizations. Scientific abstracts describing SELLAS’ findings above will be submitted for presentation in upcoming major medical meetings.

"The results of these immunodynamics assays denote a robust immunobiological foundation for the clinical effect of WT1-targeting active immunization with galinpepimut-S, and suggests key roles of both enduring CD4 activation and emergence of cross-epitopic reactivity for antimyeloma activity with this therapy. The data also support our rationale for planning further studies to expand on our experience with this IO agent," said Guenther Koehne , MD, PhD, Principal Investigator on the trial and Attending Physician, Adult Bone Marrow Transplantation Service at Memorial Sloan Kettering Cancer Center , Associate Professor of Medicine, Weill Cornell Medical College .

Dr. Nicholas Sarlis , MD, PhD, Chief Medical Officer of SELLAS, commented: "The correlations between IR and depth of clinical responses in multiple myeloma patients are both intriguing and original, and firmly establish WT1 as a major actionable target for immunotherapy in plasma cell dyscrasias henceforth."

Dr. Angelos Stergiou , MD, ScD h.c., Vice Chairman and Chief Executive Officer of SELLAS, said: "This new dataset focusing on dissecting the immune mechanisms underlying the marked antimyeloma activity previously documented with galinpepimut-S underscores our steadfast commitment to advance this agent’s clinical development programs, especially in clinical settings of unmet medical need, such as high-risk MRD(+) multiple myeloma after front-line therapy, a space which is currently underserved by standard therapies. These findings further corroborate immune response data from our earlier programs in acute myeloid leukemia (AML) and mesothelioma, where high rates of WT1-specific immunization were seen in conjunction to promising clinical activity signals in these tumor types. Finally, they provide justification for pursuance of future combinatorial approaches with galinpepimut-S, for example with checkpoint inhibitors, especially given the agent’s overall excellent safety profile."