Otsuka’s U.S. Subsidiary Astex Pharmaceuticals Enters Clinical Trial Collaboration to Explore the Potential of Combining Guadecitabine (SGI-110) with Atezolizumab in the Treatment of Acute Myeloid Leukemia

On April 19, 2016 Astex Pharmaceuticals, Inc., a pharmaceutical company dedicated to the development of novel small molecule oncology therapeutics, reported that it has entered into a clinical collaboration with Genentech (Press release, Otsuka, APR 19, 2016, View Source;date=2016-04-20 [SID:1234511123]).

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 collaboration will evaluate the potential for combining Astex’s next-generation hypomethylating agent, guadecitabine (SGI-110), with Genentech’s investigational anti-PD-L1 monoclonal antibody, atezolizumab, in the treatment of acute myeloid leukemia (AML). An initial Phase 1b study will investigate the safety and pharmacology of the combination.

OncoSec Presents Positive Melanoma Clinical Data at American Association for Cancer Research (AACR) Annual Meeting 2016

On April 19, 2016 OncoSec Medical Incorporated ("OncoSec") (NASDAQ: ONCS), a company developing DNA-based intratumoral cancer immunotherapies, today presented long-term, follow-up data of patients who were treated with its investigational therapy ImmunoPulse IL-12 and later went on to receive an anti-PD-1/PD-L1 therapy (Press release, OncoSec Medical, APR 19, 2016, View Source [SID:1234511122]). These data suggest that ImmunoPulse IL-12 may prime and enhance response rates to PD-1/PD-L1 blockade. Alain Algazi, MD, skin cancer specialist in the Melanoma Center at the UCSF Helen Diller Family Comprehensive Cancer Center, presented the findings in an oral presentation entitled "Intratumoral electroporation of plasmid IL-12 can prime response to anti-PD1/PD-L1 blockade in patients with Stage III/IV-M1a melanoma" (Abstract #CT134) at the American Association of Cancer Research (AACR) (Free AACR Whitepaper) Annual Meeting in New Orleans, LA.

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 encouraged by the data from this analysis, which show that intratumoral IL-12 DNA with electroporation can prime the immune system and help improve patient response to anti-PD-1," said Dr. Algazi. "These results are being validated prospectively in a Phase II clinical trial and they could make a clinically meaningful impact on patient outcomes and address a great unmet need in immuno-oncology."

These new data were generated from a single-site retrospective analysis of the Company’s Phase II monotherapy clinical study of ImmunoPulse IL-12, which employs intratumoral electroporation to enhance delivery of DNA-based interleukin-12 (IL-12), in patients with advanced melanoma. After completing treatment with ImmunoPulse IL-12, a subset of patients subsequently received an anti-PD-1/PD-L1 therapy either as their next line of treatment or a later line of treatment. Patients with documented follow-up history and evaluable for anti-PD-1/PD-L1 response were included in this analysis.

In this study, 34 patients were enrolled and treated with ImmunoPulse IL-12 alone. Fourteen of these 34 patients went on to receive a systemic anti-PD-1/PD-L1 therapy and were evaluable for PD-1/PD-L1 overall response rate ("ORR") using immune-related response criteria. The PD-1/PD-L1-associated ORR among patients was 64% (9/14). The analysis showed 36% of patients (5/14) had a complete response (CR), 29% of patients (4/14) had a partial response (PR), 14% percent of patients (2/14) experienced stable disease, and 21% of patients (3/14) had progressive disease. Furthermore, 8 of these 14 evaluable patients received a systemic anti-PD-1/PD-L1 antibody with no intervening therapy after treatment with ImmunoPulse IL-12. Of these 8 patients, an ORR of 75% was observed (50% CR and 25% PR).

Additionally, multiple biomarker analyses demonstrate that ImmunoPulse IL-12 therapy promotes the generation of activated natural killer and functional T cell immune subsets in the periphery as well as CD8+ tumor infiltrating lymphocytes (TIL), which may help trigger the PD-1 immune checkpoint (i.e. "adaptive immune resistance") to provide the "substrate" for effective anti-PD-1/PD-L1 therapy.

"Although one always needs to be cautious regarding the interpretation of retrospective analyses, these data are consistent with our hypothesis that ImmunoPulse IL-12 is driving a specific anti-tumor TIL response, which primes the patient for an enhanced response to PD-1 blockade," said Robert H. Pierce, MD, Chief Scientific Officer. "We look forward to following up on these observations with interim data from our ongoing combination trial in patients with melanoma investigating ImmunoPulse IL-12 and the anti-PD-1 therapy, pembrolizumab, later this year."

The full-text abstract is available and can be viewed on AACR (Free AACR Whitepaper)’s website at www.aacr.org. The presentation is available in the Publications section of OncoSec’s website.

About Melanoma
Melanoma is one of the most dangerous forms of skin cancer and accounts for the vast majority of skin cancer deaths.1 When melanoma is caught early enough, surgical excision can be curative in the majority of Stage I and II melanomas. The overall 5-year survival rate for patients with localized melanoma is 98% in the United States.1 At later stages, malignant melanoma remains a deadly and frequently difficult to treat cancer. The overall 5-year survival rate for patients falls to 17% when the disease metastasizes to distant sites or organs.1 Approximately 8,780 patients are diagnosed with Stage III and IV melanoma in the United States each year.2

Melanoma that has spread to distant sites may be treated with surgery, immunotherapy, chemotherapy and/or radiation therapy.1 Numerous chemotherapy regimens have been tested in melanoma with only modest success and limited overall survival benefit.3 Immunotherapies, such as checkpoint inhibitors, have demonstrated improvement in overall survival of patients compared to chemotherapy.3

While immunotherapy can be extremely effective, the majority of patients will not respond to anti-PD-1 therapy alone, representing a great unmet need in oncology. However, researchers are focusing efforts on targeting pathways of T cell activation.4 The presence of CD8+ T cells seems to correlate with improved prognosis and long-term survival in solid malignancies, such as melanoma,5,6 thus many emerging experimental immunotherapies seek to enhance the tumor’s immunogenicity and increase the anti-tumor CD8+ T cell response.

Kite Pharma Presents Updated Phase 1 Results from ZUMA-1 at the American Association of Cancer Research (AACR) Annual Meeting

On April 19, 2016 Kite Pharma, Inc. (Nasdaq:KITE) ("Kite") reported updated clinical results from the phase 1 portion of Kite’s ZUMA-1 trial of its lead product candidate, KTE-C19, in patients with chemorefractory, aggressive non-Hodgkin lymphoma (NHL) (Press release, Kite Pharma, APR 19, 2016, View Source [SID:1234511121]). KTE-C19 is an investigational therapy in which a patient’s T cells are genetically modified to express a chimeric antigen receptor (CAR) that is designed to target the antigen CD19, a protein expressed on the cell surface of B cell lymphomas and leukemias.

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!

David Chang, M.D., Ph.D., Kite’s Executive Vice President, Research and Development, and Chief Medical Officer, commented, "Today’s report affirms the early safety and efficacy profile of KTE-C19 in chemorefractory, aggressive NHL. We are encouraged by the ongoing complete remissions in patients with significant unmet need for new therapies. We remain on track to provide interim data from the pivotal phase 2 portion of the study later this year and to submit the KTE-C19 registration filing to the U.S. Food and Drug Administration (FDA) by the end of 2016."

"The data reported today are important because refractory DLBCL is incurable. Median survival for these patients is short and there is no standard therapy," noted Ronald Levy, M.D., Robert K. Summy and Helen K. Summy Professor of Medicine and Director of the Lymphoma Program at Stanford University School of Medicine and Associate Director of Translational Science for the Stanford Cancer Institute. "A rate of complete response and durability of response in the ranges of those reported today would be of profound clinical importance if replicated in the phase 2 portion of the ZUMA-1 study. Adoptive transfer of engineered T cells has the potential to become standard of care for patients with refractory NHL in the near future."

Updated Phase 1 Results from ZUMA-1: A Phase 1-2 Multi-Center Study Evaluating the Safety and Efficacy of KTE-C19 (Anti-CD19 CAR T Cells) in Subjects with Refractory Aggressive Non-Hodgkin Lymphoma (NHL)

Session: Early Clinical Trials Evaluating Cell-based, Checkpoint Inhibitors, and Novel Immunotherapeutics; Abstract Number: CT135; Presenter: Armin Ghobadi, M.D., Washington University, St. Louis, MO

Phase 1 of ZUMA-1 treated a total of 7 patients with chemorefractory, diffuse large B cell lymphoma (DLBCL)
KTE-C19 related adverse events consisted predominantly of cytokine release syndrome (CRS) and neurotoxicity which were generally reversible
Grade 3 or higher CRS was observed in 14% and neurotoxicity in 57%; all were reversible except in one patient with dose-limiting toxicity
KTE-C19 achieved rapid and durable responses in patients with chemorefractory disease (objective response rate 71%, complete response rate 57%)
Ongoing complete response (CR) observed in 3 of 7 patients. One ongoing CR as of 9-month study follow-up and 2 ongoing CRs as of 6-month study follow-up.
In addition, two posters on KTE-C19 engineered cell manufacturing and characteristics were presented at AACR (Free AACR Whitepaper) on April 18, 2016.

Manufacturing and Characterization of KTE-C19 in a Multicenter Trial of Patients with Refractory Aggressive Non-Hodgkin Lymphoma (NHL) (ZUMA-1)

Session: Adoptive Cell Therapy; Abstract Number: 2308; Presenter: John Rossi, M.S., Kite Pharma

The optimized GMP-manufacturing process generated anti-CD19 CAR T cells rapidly and without the need for pre-selection of a defined composition of T cells
Biologically active anti-CD19 CAR T cells were manufactured for all patients enrolled in the multicenter phase 1 ZUMA-1 trial.
Comparative Evaluation of Peripheral Blood T Cells and Resultant Engineered Anti-CD19 CAR T Cell Products from Relapsed/Refractory Non-Hodgkin’s Lymphoma (NHL) Patients

Session: Adoptive Cell Therapy; Abstract Number: 2305; Presenter: Timothy J. Langer, Kite Pharma

CAR T cells were successfully manufactured for all patients enrolled in the study at the National Cancer Institute, Surgery Branch
CAR T cell products were composed of both CD4+ and CD8+ T cells with a less differentiated phenotype than the starting leukapheresis products
CAR T cells were polyfunctional and produced a wide range of immune homeostatic, modulating and effector cytokines/chemokines in response to antigen-positive target cells.

Preclinical Data Presented At AACR Indicate Pacritinib’s Potential To Eradicate Therapy-Resistant Leukemia Stem Cells Residing In Bone Marrow Microenvironment

On April 20, 2016 CTI BioPharma Corp. (CTI) (NASDAQ and MTA:CTIC) reported findings from an investigator-sponsored preclinical study indicating that pacritinib, an inhibitor of JAK2, FLT3, IRAK1 and CSF1R, may be effective in reducing survival of myelofibrosis and acute myeloid leukemia (AML) repopulating cells (Press release, CTI BioPharma, APR 19, 2016, View Source;p=RssLanding&cat=news&id=2158522 [SID:1234511114]). Further, this study also demonstrated that the combination of pacritinib at low nanomolar concentrations with dasatinib may eliminate self-renewing leukemia stem cells in blast crisis of chronic myeloid leukemia (CML) with minimal toxicity toward normal progenitors. In myeloid leukemias, these leukemic stem cells can evade initial treatment and hide within the bone marrow microenvironment, develop resistance to current therapies, self-renew and eventually cause relapse.

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!

These findings were presented by Larissa Balaian, Ph.D. from the Moores Cancer Center, University of California San Diego in a poster presentation (abstract #3338) titled: "Pacritinib reduces human myeloid leukemia stem cell maintenance in a defined niche," during the American Association of Cancer Research (AACR) (Free AACR Whitepaper) Annual Meeting held April 16-20 in New Orleans, LA.

"The potential ability for pacritinib to eradicate therapy resistant leukemia stem cells in relapse AML as a single-agent, as well as eliminate self-renewing stem cells in CML, when used in combination with standard of care therapy, demonstrates that targeting niche-dependent signaling with pacritinib could represent a new approach to treating patients with refractory acute myeloid leukemia and blast crisis of CML," said Dr. Balaian.

Additional data being presented at the meeting include:

A poster (abstract #2602) titled: "The nonclinical toxicology profile of pacritinib, a JAK2/FLT3 inhibitor with no dose-limiting clinical myelosuppression." In this poster, CTI BioPharma researchers presented data from studies of pacrinitib in nonclinical models that were evaluated in comparison to publicly available information for the currently approved JAK inhibitors. The nonclinical toxicology profile findings showed that pacritinib is unique for its mild myelosuppressive effects in the nonclinical studies. Of interest, only pacritinib was not associated with increased opportunistic infections in the long-term toxicology studies.

A poster (abstract #1609) titled: "Investigation of absorption, metabolism, excretion, and mass balance of [14C]-pacritinib in healthy subjects: a phase 1 study." In this poster, CTI BioPharma researchers investigated clearance pathways, excretion, pharmacokinetics and recovery of pacritinib’s major metabolites in healthy volunteers. Intact pacritinib was minimally excreted in urine and feces while most radioactivity was recovered as metabolites in feces, suggesting extensive biliary clearance and hepatic metabolism of pacritinib. No dose adjustments are anticipated to be required for patients with renal impairment.

The foregoing summaries of such reported findings and posters are not complete and are qualified in their entirety by reference to the referenced posters. These and other poster presentations are available in the publication section of the CTI BioPharma website at ctibiopharma.com.

About Pacritinib

Pacritinib is an investigational oral kinase inhibitor with specificity for JAK2, FLT3, IRAK1 and CSF1R. In August 2014, pacritinib was granted Fast Track designation by the FDA for the treatment of intermediate and high risk myelofibrosis including, but not limited to, patients with disease-related thrombocytopenia (low platelet counts); patients experiencing treatment-emergent thrombocytopenia on other JAK2 inhibitor therapy; or patients who are intolerant of, or whose symptoms are not well controlled (sub-optimally managed) on other JAK2 therapy. Clinical studies for pacritinib are currently subject to a full clinical hold issued by the U.S. Food and Drug Administration in February 2016. The Company is in the process of responding to the full clinical hold by working through the FDA’s recommendations prior to requesting a meeting with them. In March 2016, the FDA expressed interest in allowing patients who were receiving benefit from pacritinib treatment at the time the clinical hold was imposed to submit requests to the FDA to resume pacritinib treatment under a Single Patient IND (SPI) program on a case-by-case basis. The Company is working with investigators in submitting SPI requests to the FDA. Separately, the FDA has informed clinical investigators that emergency requests may be submitted to the FDA for individual patient Expanded Access to pacritinib. Expanded Access, sometimes called "compassionate use," is the use outside of a clinical trial of an investigational medical product. Pacritinib does not have regulatory approval and is not commercially available.

CTI BioPharma and Baxalta Incorporated are parties to a worldwide license agreement to develop and commercialize pacritinib. CTI BioPharma and Baxalta will jointly commercialize pacritinib in the U.S., while Baxalta has exclusive commercialization rights for all indications outside the U.S.

Peloton Therapeutics, Inc. Presents Positive Preclinical Data on First HIF-2α Inhibitor in Combination with Immuno-oncology Agents at 2016 AACR Annual Meeting

On April 19, 2016 Peloton Therapeutics, Inc., a drug discovery and development company focused on advancing first-in-class, small molecule cancer therapies targeting unexploited molecular vulnerabilities, reported preclinical data on its lead investigational candidate, PT2385, at the American Association for Cancer Research (AACR) (Free AACR Whitepaper) Annual Meeting in New Orleans, LA (Press release, Peloton Therapeutics, APR 19, 2016, View Source [SID:1234511099]). PT2385 is the first clinical stage antagonist of hypoxia inducible factor-2α (HIF-2α), a transcription factor implicated in the development and progression of renal and other cancers.

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!

"PT2385, a Novel HIF-2α Antagonist, Combines with Checkpoint Inhibitor Antibodies to Inhibit Tumor Growth in Preclinical Models by Modulating Myeloid Cells and Enhancing T Cell Infiltration"
Tweet this
In addition to its direct role in transcription regulation of growth-promoting genes in renal tumors, HIF-2α has been proposed to affect the tumor microenvironment. In a poster titled "PT2385, a Novel HIF-2α Antagonist, Combines with Checkpoint Inhibitor Antibodies to Inhibit Tumor Growth in Preclinical Models by Modulating Myeloid Cells and Enhancing T Cell Infiltration," the combination of PT2385 with antibodies to immune checkpoint control molecules (PD-1, PD-L1, and CTLA4) yielded additive or synergistic efficacy in preclinical tumor models. HIF-2α is not detected in the mouse tumor cells, but is expressed in the stroma. Tumor growth inhibition by these combination regimens was accompanied by modulation of a variety of immune markers such as infiltrating T-cells, macrophage and myeloid-derived suppressor cell populations in the tumors. The combination of PT2385 and immune checkpoint inhibitors is planned for evaluation in clinical trials.

"PT2385 has now been shown to affect the tumor microenvironment, even for tumors that do not express HIF-2α. This potentially broadens the applicability of PT2385 to a larger variety of tumor types, including melanoma and lung cancers, which have been shown to have a strong immunological component," said John Josey, Ph.D., Peloton’s Chief Executive Officer.

About PT2385

PT2385 is a first-in-class small molecule inhibitor of hypoxia-inducible factor-2α (HIF-2α), a transcription factor implicated in the development and progression of kidney cancer. It is currently being investigated in a Phase 1 clinical trial for the treatment of advanced or metastatic clear cell renal cell carcinoma (ccRCC). Loss of the von Hippel-Lindau tumor suppressor (VHL) is the key oncogenic event in up to 95% of patients with ccRCC. With the loss of the VHL protein (pVHL), the transcription factor HIF-2α accumulates and drives the unbalanced expression of numerous gene products. Preclinical data indicate that orally bioavailable PT2385 disrupts HIF-2α activity in ccRCC and thereby blocks the expression of multiple tumorigenic factors responsible for unrestrained cancer cell growth and proliferation, tumor angiogenesis, and suppression of anti-tumor immune responses characteristic of ccRCC.

About Renal Cell Cancer

The American Cancer Society estimates that more than 62,000 new cases of kidney cancer will be diagnosed and more than 14,000 people will die from this disease this year. The National Cancer Institute reports that the prognosis for any treated renal cell cancer patient with progressing, recurring, or relapsing disease is poor, regardless of cell type or stage.