Pipeline Review Check

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!

MT-3724 1st-Generation Forced Internalization Novel MOA CD20 B-Cell Malignancies CD20 Internalization Novel MOA

79

MT-4019 2nd-Generation
De-immunized CD38 Multiple Myeloma Active on CD38 low, CD55/CD59 high tumor cells

42

MT-5111 2nd-Generation
De-immunized HER2 Breast Cancer Novel epitope/combinable with Kadcyla Novel MoA
Active in Kadcyla-refractory cells

37

MT-5050 PD-L1+ tumors PD-L1 Melanoma Novel MoA Antigen seeding

25

MT-6868 1st-Generation CD45 SCT Novel MOA

Bristol-Myers Squibb Announces Topline Results from CheckMate -214, a Phase 3 Study of Opdivo in Combination with Yervoy in Intermediate and Poor-Risk Patients with Previously Untreated Advanced or Metastatic Renal Cell Carcinoma

On August 15, 2017 Bristol-Myers Squibb Company (NYSE: BMY) reported topline results today from the CheckMate -214 trial investigating Opdivo (nivolumab) in combination with Yervoy (ipilimumab) versus sunitinib in intermediate and poor-risk patients previously untreated advanced or metastatic renal cell carcinoma (Press release, Bristol-Myers Squibb, AUG 15, 2017, View Source [SID1234520256]).

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 combination met the co-primary endpoint of objective response rate (ORR) and achieved a 41.6% ORR versus 26.5% for sunitinib. Median duration of response was not reached for the combination of Opdivo and Yervoy and was 18.17 months for sunitinib. While there was an improvement in progression-free survival (PFS) (HR=0.82, [95% CI 0.64 – 1.05]; stratified 2-sided p=0.03), it did not reach statistical significance. The median PFS was 11.56 months (95% CI 8.71 – 15.51) for the Opdivo and Yervoy combination versus 8.38 months (95% CI 7.03-10.81) for sunitinib, The study will continue as planned to allow the third co-primary endpoint of overall survival to mature. The tolerability profile observed in CheckMate-214 was consistent with that observed in previously reported studies of this dosing schedule.

"We are encouraged by the totality of the CheckMate-214 data. The overall response rate and durability of response favored the combination of Opdivo and Yervoy, and the trend for PFS supports the potential of the combination in intermediate and poor-risk advanced renal cell carcinoma, the most common type of kidney cancer. This is an important study in first-line renal cancer as these patients need new options," said Vicki Goodman, M.D., development lead, Melanoma and Genitourinary Cancers, Bristol-Myers Squibb. "We look forward to presenting the full results from this study at an upcoming medical meeting."

"Opdivo is now well established as standard of care in the treatment of second-line renal cell carcinoma," said Goodman. "The company plans to share these important first-line data with regulatory authorities and we look forward to reporting overall survival when these data mature."

About CheckMate -214

CheckMate -214 is a phase 3, randomized, open-label study evaluating the combination of Opdivo plus Yervoy versus sunitinib in patients with previously untreated advanced or metastatic renal cell carcinoma. Patients in the combination group received Opdivo 3 mg/kg plus Yervoy 1 mg/kg every 3 weeks for 4 doses followed by Opdivo 3 mg/kg every 2 weeks. Patients in the comparator group received sunitinib 50 mg once daily for 4 weeks, followed by 2 weeks off before continuation of treatment. Patients were treated until progression or unacceptable toxic effects. The primary endpoints of the trial are progression-free survival, overall survival and objective response rate in an intermediate to poor-risk patient population (approximately 75 percent of patients). The majority of alpha was allocated to overall survival. Safety is a secondary endpoint.

About Renal Cell Carcinoma

Renal cell carcinoma (RCC) is the most common type of kidney cancer in adults, accounting for more than 100,000 deaths worldwide each year. Clear-cell RCC is the most prevalent type of RCC and constitutes 80% to 90% of all cases. RCC is approximately twice as common in men as in women, with the highest rates of the disease in North America and Europe. Globally, the five-year survival rate for those diagnosed with metastatic, or advanced kidney cancer, is 12.1%.

VBI Vaccines Announces FDA Acceptance of Investigational New Drug Application for VBI-1901 to Treat Glioblastoma Multiforme

On August 15, 2017 VBI Vaccines Inc. (Nasdaq: VBIV) (TSX: VBV) (VBI), a commercial-stage biopharmaceutical company developing next-generation infectious disease and immuno-oncology vaccines, reported that the U.S. Food and Drug Administration (FDA) has accepted the company’s Investigational New Drug Application (IND) for VBI-1901, a novel immunotherapy targeting Glioblastoma Multiforme (GBM), one of the most common and aggressive malignant primary brain tumors in humans (Press release, VBI Vaccines, AUG 15, 2017, View Source [SID1234520255]). The IND enables VBI to initiate a multi-center Phase I/2a clinical study evaluating VBI-1901 in patients with recurrent GBM in the second half of this year.

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!

VBI has leveraged its enveloped virus-like particle (eVLP) platform and expertise in immunology to develop a broadly active therapeutic vaccine that targets two highly immunogenic CMV antigens, gB and pp65, to direct a potent immune response against CMV infection. Scientific literature suggests CMV infection is prevalent in multiple solid tumors, including GBM. The vaccine candidate is combined with granulocyte-macrophage colony-stimulating factor (GM-CSF), an adjuvant that mobilizes dendritic cell function and seeks to enhance productive immunity against tumors.

"A growing body of research has demonstrated that GBM tumors may be susceptible to infection by CMV, with over 90% of GBM tumors expressing CMV antigens," said Jeff Baxter, president and CEO of VBI. "FDA clearance of this IND is a significant milestone for VBI as we expand our eVLP platform into immuno-oncology applications."

GBM Program Background

Glioblastoma is among the most common and aggressive malignant primary brain tumors in humans. In the U.S. alone, 12,000 new cases are diagnosed each year. The current standard of care for treating GBM is surgical resection, followed by radiation and chemotherapy. Even with aggressive treatment, GBM progresses rapidly and is exceptionally lethal, with median patient survival of less than 16 months.

Targeted immunotherapy may provide a promising adjunct or alternative to conventional GBM treatment. Immunotherapy is a fundamentally different way of treating cancer that stimulates the patient’s immune system to resume its attack on tumors. While conventional therapies are non-specific and may damage surrounding normal tissues, targeted immunotherapy may offer a highly specific and potentially long-lasting treatment approach that leverages the immune system to protect against cancer.

Developing a broadly applicable GBM immunotherapy requires the identification of antigens that are consistently expressed on GBM tumor cells. Recent research has demonstrated that an anti-CMV dendritic cell vaccination regimen may extend overall survival in patients with GBM. Thus, effective targeting of CMV antigens may represent an attractive strategy for a GBM immunotherapy.

Sierra Oncology Establishes Distinguished DNA Damage Response Advisory Committee

On August 15, 2017 Sierra Oncology, Inc. (NASDAQ: SRRA), a clinical stage drug development company focused on advancing next generation DNA Damage Response (DDR) therapeutics for the treatment of patients with cancer, reported it has established a DDR Advisory Committee composed of leading experts in this emerging field of cancer drug development (Press release, ProNAi Therapeutics, AUG 15, 2017, View Source [SID1234520254]). The DDR Advisory Committee will advise Sierra’s management as it advances its DDR oriented development programs and seeks to maximize the potential clinical and commercial deployment of its drug candidates.

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!

"A growing body of research has established that the DDR network is essential for cancer cell survival and adaptation. As such it presents fertile ground for generating innovative approaches to treat various cancers. To capitalize on this rapidly advancing and evolving science, we are collaborating closely with Cancer Research UK and the Institute of Cancer Research, institutions that are pioneers in DDR and the originators of our lead DDR asset, SRA737, which targets Chk1, a critical DDR regulator," said Dr. Nick Glover, President and CEO of Sierra Oncology. "We believe the formation of our new DDR Advisory Committee, represented by leading experts in DDR biology, chemistry and medicine, will further strengthen our ability to remain at the forefront of this field and demonstrates our commitment to this promising new area in cancer treatment."

Sierra Oncology’s DDR Advisory Committee consists of:
Eric J. Brown, PhD, Associate Professor of Cancer Biology at the Perelman School of Medicine of the University of Pennsylvania.

Karlene Cimprich, PhD, Vice Chair and Professor of Chemical and Systems Biology at the Stanford University School of Medicine.

Alan D. D’Andrea, MD, Fuller-American Cancer Society Professor of Radiation Oncology at Harvard Medical School and the Director of the Center for DNA Damage and Repair at the Dana-Farber Cancer Institute.

Alan R. Eastman, PhD, Professor at the Geisel School of Medicine at Dartmouth and the founding Director of the Molecular Therapeutics Research Program of the Norris Cotton Cancer Center at Dartmouth.

Michelle D. Garrett, PhD, Professor of Cancer Therapeutics in the School of Biosciences at the University of Kent and Visiting Professor of Cancer Therapeutics at the Institute of Cancer Research, London, UK.

Thomas Helleday, PhD, The Torsten and Ragnar Söderberg Professor of Translational Medicine and Chemical Biology at Karolinska Institutet, Stockholm, Sweden.

Leonard Post, PhD, Chief Scientific Officer of Vivace Therapeutics.
"We aim to maintain a leading position in the clinical translation of DDR science and view this team as integral to that objective. Members of our DDR Advisory Committee were selected to add specific expertise that will provide the company with complementary, well-rounded guidance and strategic advice," added Dr. Christian Hassig, Senior Vice President, Research of Sierra Oncology. "As we advance our pipeline, a thorough understanding of emerging DDR biology will inform our clinical strategies and help maximize the potential clinical opportunities for our assets."

About Sierra’s DDR Advisory Committee members:
Eric J. Brown, PhD, Associate Professor of Cancer Biology at the Perelman School of Medicine at the University of Pennsylvania.

Dr. Brown’s laboratory at the University of Pennsylvania examines how signaling maintains genome stability during DNA synthesis and how this function is essential to cancer cells. His laboratory was the first to report that oncogenic stress is sufficient to cause selective sensitivity to ATR inhibition. Dr. Brown’s laboratory is currently identifying predictive biomarkers of therapeutic benefit and the mechanisms of action of these drugs through a combination of genome-wide breakpoint mapping and replication fork proteomics. In collaboration with clinical researchers, these biomarkers of response will be exploited in current and future clinical trials. Collectively, the Brown laboratory seeks both to define the mechanisms of action of ATR/Chk1 inhibitors and to identify their optimal uses in cancer therapies.

Dr. Brown received his BA (Genetics) from the University of California at Berkeley (1989) and his PhD (Immunology)
from Harvard University (1996). He performed his doctoral research with Dr. Stuart Schreiber at Harvard University, where he purified and cloned the mammalian target of rapamycin (mTOR). In his postdoctoral research in Dr. David Baltimore’s laboratory at the California Institute of Technology, Dr. Brown investigated the impact of ATR suppression on genome stability and checkpoint signaling in response to replication stress.

Karlene Cimprich, PhD, Vice Chair and Professor, Department of Chemical and Systems Biology at the Stanford University School of Medicine.

The Cimprich lab is focused on understanding how cells maintain genomic stability, with an emphasis on the DNA damage response. These include effects on DNA repair, transcription, and DNA replication, as well as cell cycle arrest, apoptosis, and senescence. The lab is particularly interested in understanding how DNA damage is identified and resolved during DNA replication, when the genome is particularly vulnerable due to stalling of the replication fork at naturally arising and induced DNA lesions, structures or protein-DNA complexes. Dr. Cimprich’s research has evaluated the role of proteins, including Chk1 and Cdc7, in replication fork collapse.

Dr. Cimprich received her BS, from the University of Notre Dame, Chemistry (1989) and her PhD from Harvard University, Chemistry (1994). She was a Postdoctoral Fellow in the Department of Chemistry and Chemical Biology, Harvard University in the laboratory of Dr. Stuart Schreiber. Dr. Cimprich is a AAAS fellow and a recipient of the Kimmel Scholar Award, Burroughs Wellcome New Investigator Award, and the Ellison Senior Scholar Award.
Alan D. D’Andrea, MD, Fuller-American Cancer Society Professor of Radiation Oncology at Harvard Medical School and the Director of the Center for DNA Damage and Repair at the Dana-Farber Cancer Institute.

Dr. D’Andrea is internationally known for his research in the area of DNA damage and DNA repair. Through his work on DNA repair biomarkers, Dr. D’Andrea participates in a wide range of clinical trials, largely focused on ovarian, breast, prostate, and bladder cancers. His research has focused on the molecular cause of leukemia for many years and he also investigates the pathogenesis of Fanconi anemia DNA repair pathway, a human genetic disease characterized by bone marrow failure and AML in children. Dr. D’Andrea has previously published on the correlation between DNA repair deficient pathways and Chk1 inhibition.

Dr. D’Andrea received his MD from Harvard Medical School in 1983. He completed his residency in Pediatrics at Children’s Hospital of Philadelphia, and a fellowship in hematology-oncology at DFCI and Children’s Hospital, Boston. Dr. D’Andrea also completed a research fellowship at the Whitehead Institute of Biomedical Research at MIT where he cloned the receptor for erythropoietin while working in the laboratory of Harvey Lodish. Dr. D’Andrea joined the staff at DFCI in 1990 and in 2017 he became the Director of the Susan Smith Center for Women’s Cancer at the DFCI. A recipient of numerous academic awards, Dr. D’Andrea is a former Stohlman Scholar of the Leukemia and Lymphoma Society, and serves on their Medical and Scientific Advisory Board.

Alan R. Eastman, PhD, Professor at the Geisel School of Medicine at Dartmouth and the founding Director of the Molecular Therapeutics Research Program of the Norris Cotton Cancer Center at Dartmouth.

Dr. Eastman’s research has concentrated in the area of preclinical cancer chemotherapy through early phase clinical trials. His initial research focused primarily on the mechanism of action of cisplatin which led to analysis of the resulting cell cycle perturbation and the identification of apoptosis as an end point. These two areas, apoptosis and cell cycle checkpoint regulation, continue today with the overall goal of bringing new drugs and new strategies to clinical trials. Dr. Eastman has published extensively on Chk1 inhibition as well as Cdc7 inhibition and their respective mechanisms of action. Further, Dr. Eastman has participated in Chk1 inhibitor clinical trials.

Dr. Eastman received his BTech from Brunel University, London (1972) and his PhD from the Chester Beatty Research Institute, University of London (1975). In 1989, he joined the faculty at the Geisel School of Medicine at Dartmouth where he has been a professor since 1992. In 1993, he was the founding Director of the Molecular Therapeutics Research Program of the Norris Cotton Cancer Center at Dartmouth, an NCI-designated comprehensive cancer center, and he continues in this role today. Prior to this, he took a position in 1983 as associate professor at the Eppley Institute for Research in Cancer, University of Nebraska Medical Center.

Michelle D. Garrett, PhD, Professor of Cancer Therapeutics in the School of Biosciences at the University of Kent and Visiting Professor of Cancer Therapeutics at the Institute of Cancer Research, London, UK.

Dr. Garrett’s current research is focused on understanding the molecular and cellular effects of drugs that target cell signaling pathways and the cell division cycle. This research includes Chk1 inhibitors such as SRA737 and other functionally related drugs. One of the long-term aims of this work is to identify biomarkers for both patient stratification and drug-target engagement for these drugs in the clinic. Dr. Garrett is a co-inventor of SRA737 and related compounds.

Dr. Garrett received her undergraduate degree from the University of Leeds, UK (1987) and her PhD from The Institute of Cancer Research (ICR), London, UK (1991). She undertook post-doctoral research at Yale Medical School, USA and then in 1994 joined Onyx Pharmaceuticals, California, USA, where she was involved in the discovery of small molecule drugs targeting the cell cycle. This included the CDK4 inhibitor project, which went on to deliver Ibrance (Palbociclib) now a registered treatment for breast cancer. In 1999, Dr. Garrett returned to the ICR where she went on to become a Reader in Cancer Therapeutics and Head of Biology for the CRUK Cancer Therapeutics Unit. In September 2014, she joined the School of Biosciences at the University of Kent and currently has three cancer drugs in the clinic.

Thomas Helleday, PhD, The Torsten and Ragnar Söderberg Professor of Translational Medicine and Chemical Biology at Karolinska Institutet, Stockholm, Sweden.

Professor Helleday heads a large multidisciplinary translational research group at Karolinska Institute focusing on understanding basic DNA repair and DNA-damage and developing novel drugs for anti-cancer treatments. The group was first to demonstrate a novel concept for treating cancer called "synthetic lethality" established by the selective killing of BRCA1 or BRCA2 mutated breast and ovarian cancers by PARP inhibitors (now EMA/FDA approved). The research covers both basic and translational work including academic-driven clinical trials, based on basic science findings in his laboratory. Dr. Helleday has published extensively on the ATR/Chk1 pathway, and has established a cancer specific synthetic lethal relationship between ATR and Chk1 inhibition.

Professor Helleday received a degree in molecular biology (1995) and in Business Administration and Economics (1996) from the Stockholm University. He received a PhD from Stockholm University in 1999 for his studies on homologous recombination in mammalian cells. Professor Helleday has been awarded numerous eminent international grants and awards in recognition of his research accomplishments including the Eppendorf-Nature Young European Investigator Award (2005) for outstanding contribution within the field of biomedical science by the journal Nature and two prestigious ERC advanced grants (2010 and 2016).

Leonard Post, PhD, Chief Scientific Officer of Vivace Therapeutics.
Dr. Post is Chief Scientific Officer of Vivace Therapeutics and also serves as an advisor to numerous biotechnology companies and to venture investors. Until July 2016, he was Chief Scientific Officer of BioMarin Pharmaceuticals, and before that was CSO and co-founder of LEAD Therapeutics which was acquired by BioMarin in 2010. His work in DNA repair involved the discovery of the PARP inhibitor talazoparib at LEAD and its development into Phase 3 at BioMarin.

Talazoparib is currently being tested in EMBRACA, a Phase III clinical study in gBRCA+ locally advanced and/or metastatic breast cancer. From 2000-2006, he was Senior Vice President of Research and Development at Onyx Pharmaceuticals, during the clinical development of Nexavar from IND through NDA approval. Prior to Onyx, he was at Parke-Davis Pharmaceutical where he was VP of Discovery Research; and before that at The Upjohn Company in several positions. Dr. Post is currently a member of the board of directors of Viralytics, an ASX-listed company; and of private companies Orphagen Pharmaceuticals, Fedora Pharmaceuticals and Oxyrane Ltd.

Dr. Post received a Bachelor of Science in Chemistry from the University of Michigan and a PhD in Biochemistry from the University of Wisconsin, Madison after which he performed a Postdoctoral Fellowship in Virology at the University of Chicago. From 1993 to June 2000, he served as Adjunct Professor in the Department of Microbiology and Immunology at the University of Michigan.

Onconova Therapeutics, Inc. Reports Recent Business Highlights and Second Quarter 2017 Financial Results

On August 15, 2017 Onconova Therapeutics, Inc. (NASDAQ:ONTX), a Phase 3 stage biopharmaceutical company focused on discovering and developing novel small molecule drug candidates to treat cancer, with a primary focus on Myelodysplastic Syndromes (MDS), reported a corporate update and reported financial results for the second quarter ended June 30, 2017 (Press release, Onconova, AUG 15, 2017, View Source [SID1234520247]).

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 advancing IV rigosertib in a late stage clinical trial for patients with unmet medical needs in MDS. Our Phase 3 INSPIRE trial with IV rigosertib is enrolling globally and the next milestone of interim analysis is anticipated in the fourth quarter," said Dr. Ramesh Kumar, President and Chief Executive Officer.

"After consulting with regulators in the US and Europe following Phase 1/2 data with oral rigosertib in combination with azacitidine, we are designing a Phase 3 trial in first-line higher risk MDS patients for submission under a Special Protocol Assessment," continued Dr. Kumar. "We also believe there is interest for our pediatric RASopathies rare disease collaborative program from the National Cancer Institute, academia, and patient advocacy which we find extremely gratifying."

INSPIRE Trial of IV Rigosertib in 2nd Line Higher-risk (HR) MDS

Interim Analysis

The INSPIRE protocol allows for a pre-planned interim analysis. This analysis will be triggered after reaching 88 events (deaths). Although it is difficult to accurately forecast the timing of this milestone, based on when we expect to finalize our statistical analysis plan, enrollment statistics, and our expectations for survival of the trial population, we anticipate this to occur in Q4-2017.
The Statistical Analysis Plan (SAP) for interim and top-line analysis is under review by the Food and Drug Administration (FDA) and the European Medicines Agency (EMA).
Trial Progress

As of July 31, 72 sites in 16 countries across four continents have enrolled patients for this study. Countries and trial sites were carefully chosen to ensure availability of appropriate patients meeting stringent eligibility criteria. Since these criteria were purposely designed to be narrow and selective, extensive eligibility verification and trial site education are integral to our plan.
Enrollment for the trial has slowed recently, which could be related to seasonality. The Company is taking proactive measures to increase enrollment including the addition of trial sites in three new countries and changes within its CRO group. Should enrollment not return to desired levels, full enrollment may be delayed by several months.
Oral Rigosertib in Combination with Azacitidine for 1st-line HR-MDS

Pivotal Phase 3 Trial Protocol

Following receipt of the final minutes from our End-of-Phase 2 discussion with the FDA in 3Q-2016, a Scientific Advice process was initiated with the EMA and was completed in July. Based on this feedback, we are designing a Phase 3 protocol for a 1:1 randomized controlled trial of oral rigosertib + azacitidine compared with azacitidine + placebo in first-line patients with HR-MDS. We plan to initiate the FDA Special Protocol Assessment (SPA) process following completion of the ongoing Expansion Phase 1/2 trial.
Initiation of the trial, which is planned to be conducted globally, requires additional financing.
Expansion of Phase 1/2 Trial of Oral Rigosertib in Combination with Azacitidine

This expansion phase is designed to enroll up to approximately 40 patients. The key objectives are to optimize dosing and schedule of administration of oral rigosertib in combination with azacitidine.
After appropriate amendments were filed with the regulatory agencies, we started the expansion phase of this trial. Four sites are now open in the U.S. and we plan to activate additional sites in US, Europe and Australia. The first patient was enrolled in April.
Other Programs for Future Development or Partnership and Presentations

New Collaborative Program in "RASopathies": Rare Disease in Children

Based on new mechanism of action data published last year (Cell, 2016), we initiated a collaborative development program focusing on a group of rare diseases, RASopathies, which share a well-defined molecular basis in expression or defects involving Ras Effector Pathways. Pediatric "RASopathies" is a subset of these rare diseases, and we are embarking on a multifaceted collaborative program involving patient advocacy, government, and academic organizations. In addition to drug supply and expertise, we believe we can contribute organizational oversight to this project.

We expect to execute a formal Collaborative Research and Development Agreement (CRADA) with the National Cancer Institute (NCI) of the National Institutes of Health.

Investigators in the NCI have designed a protocol for a trial in pediatric patients with certain RASopathies complicated by cancer.

Since our focus area of MDS is related to a pediatric rare disease, Juvenile Myelomonocytic Leukemia (JMML), we are exploring collaboration with an academic group focused on development of novel therapeutics for these children. Initial non-clinical studies in Stem Cells and animal models may influence the design of an appropriate clinical study in this patient population.

In July 2017, we presented a summary of our targeted approach to a symposium organized by Rasopathiesnet.org, a patient advocacy organization. We are also sponsoring an educational event (a "Key Opinion Leader" breakfast) in the fourth quarter to bring together disease area experts, patient advocacy groups and our knowledge, to draw attention to this unmet medical need and the potential for rigosertib in this quest.
Recent Presentations

The Company presented a poster at the American Society of Clinical Oncology (ASCO) (Free ASCO Whitepaper) Annual Meeting on Monday, June 5th focusing on "Further Rationale for Rigosertib in a Second-line HR-MDS Setting." Bone marrow response was evaluated as a surrogate for survival in this trial of 64 patients who had failed hypomethylating agents. 22% of these patients achieved marrow complete response (mCR) and 47% of patients achieved disease stabilization.

The Company contributed multiple presentations at the 22nd Congress of the European Hematology Association (EHA) (Free EHA Whitepaper) on June 24th. The presentations incorporated clinical and non-clinical data evaluating oral rigosertib plus azacitidine. The data presented demonstrated that the combination may overcome hypomethylating agent resistance, and also indicates that further study in acute myeloid leukemia is warranted. SymBio, Onconova’s partner in Japan and Korea, presented Phase 1 safety data for intravenous rigosertib in Japanese patients with recurrent/relapsed or refractory Myelodysplastic Syndromes.

Dr. Steven Fruchtman, Onconova’s Chief Medical Officer, highlighted approaches for studying rigosertib in RAS-associated cancers on Sunday, July 30th at the 5th International RASopathies Symposium organized by RASopathies Network. The primary focus of this presentation was JMML, which is incurable without an allogenic hematopoietic stem cell transplant.
Proprietary Preclinical Next Generation CDK4/6 + ARK5 Inhibitor

New CDK inhibitors are making an impact for patients with breast and lung cancers. These orally available agents have potential utilities in a broad range of solid tumors and we believe will become an integral part of cancer therapy in the future.

Our CDK inhibitor is differentiated from other agents on the market (such as Palbociclib, Ribociclib and Abemaciclib) or in development (such as the compounds being developed by G1 Therapeutics) by its dual inhibition of CDK4/6 + ARK5. We continue to carry out preclinical research to enhance the data package for this compound in an attempt to seek partners for co-development of this novel new chemical entity.
Second-Quarter Business Highlight:

On April 26, 2017, Onconova closed a public offering resulting in gross proceeds of approximately $5.2 million, before underwriting discounts, commissions and estimated offering costs. New institutional investors, existing investors, as well as Directors and Management of the Company participated in the round. On May 17, 2017, the underwriters exercised their option to purchase an additional 363,580 shares, which resulted in additional gross proceeds of $0.8 million.
Second-Quarter Financial Results:

Cash, cash equivalents, and marketable securities as of June 30, 2017 totaled $15.0 million, compared to $21.4 million as of December 31, 2016.

Total net revenue was $0.3 million for the second quarter of 2017 and $0.5 million for the six months ended June 30, 2017, compared to $2.2 million and $3.7 million, respectively, for the comparable periods in 2016.

Research and development expenses were $4.6 million for the second quarter of 2017 and $9.5 million for the six months ended June 30, 2017, compared to $5.6 million and $11.4 million, respectively, for the comparable periods in 2016.

General and administrative expenses were $1.8 million for the second quarter of 2017 and $3.9 million for the six months ended June 30, 2017, compared to $2.1 million and $5.3 million, respectively, for the comparable periods in 2016.

About IV Rigosertib

The intravenous form of rigosertib has been employed in Phase 1, 2, and 3 clinical trials involving more than 800 patients, and is currently being evaluated in the randomized Phase 3 international INSPIRE trial for patients with higher-risk (HR) MDS, after failure of hypomethylating agent, or HMA, therapy. This formulation is intended for patients with advanced disease, to provide long duration of exposure, and to ensure dosing under a controlled setting.

About INSPIRE

The INternational Study of Phase III IV RigosErtib, or INSPIRE, is based on guidance received from the U.S. Food and Drug Administration and European Medicines Agency and derives from the analysis of the ONTIME Phase 3 trial. INSPIRE is a multi-center, randomized controlled study to assess the efficacy and safety of IV rigosertib in HR-MDS patients who had progressed on, failed to respond to, or relapsed after previous treatment with an HMA within the first 9 months or nine cycles over the course of one year after initiation of HMA treatment. This time frame optimizes the opportunity to respond to treatment with an HMA prior to declaring treatment failure, as per the National Comprehensive Cancer Network (NCCN) Guidelines. The trial will enroll approximately 225 patients randomized at a 2:1 ratio into two treatment arms: IV rigosertib plus Best Supportive Care versus Physician’s Choice plus Best Supportive Care. The primary endpoint of INSPIRE is overall survival and an interim analysis is anticipated. Full details of the INSPIRE trial, such as inclusion and exclusion criteria, as well as secondary endpoints, can be found on clinicaltrials.gov (NCT02562443).

About Oral Rigosertib

The oral form of rigosertib was developed to provide more convenient dosing for use where the duration of treatment may extend to multiple years. This dosage form also supports many combination therapy modalities. To date, 368 patients have been treated with the oral formulation of rigosertib. Initial studies with single-agent oral rigosertib were conducted in hematological malignancies, lower-risk MDS, and solid tumors. Combination therapy of oral rigosertib with azacitidine and chemoradiotherapy has also been explored. Currently, oral rigosertib is being developed as a combination therapy together with azacitidine for patients with higher-risk MDS who require HMA therapy. A Phase 1/2 trial of the combination therapy has been fully enrolled and the preliminary results were presented in 2016. This novel combination is the subject of an issued US patent with earliest expiration in 2028.