Seattle Genetics Highlights Data Supporting Antibody-Drug Conjugate and Novel Immuno-Oncology Programs at the American Association for Cancer Research (AACR) Annual Meeting

On April 18, 2016 Seattle Genetics, Inc. (NASDAQ: SGEN) reported multiple data presentations that support several of the company’s antibody-drug conjugate (ADC) and immuno-oncology programs featured at the upcoming 107th Annual Meeting of the American Association for Cancer Research (AACR) (Free AACR Whitepaper) being held April 16 through 20, 2016 in New Orleans, LA (Press release, Seattle Genetics, APR 18, 2016, View Source;p=RssLanding&cat=news&id=2157809 [SID:1234510980]). The presentations describe preclinical data with SGN-LIV1A and SEA-CD40, which are in ongoing clinical trials, and highlight two preclinical programs, a novel ADC for the treatment of multiple myeloma (SGN-CD352A) and a small molecule that enhances T-cell mediated antitumor activity (2-fluorofucose). Additional data highlight the growing body of knowledge regarding the ability of auristatin-based ADCs, including ADCETRIS (brentuximab vedotin), to initiate immunogenic cell death, supporting evaluation in combination with checkpoint inhibitors.

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!

"Our ADC expertise currently empowers a robust pipeline of more than a dozen clinical and preclinical programs," said Jonathan Drachman, M.D., Chief Medical Officer and Executive Vice President, Research and Development at Seattle Genetics. "At AACR (Free AACR Whitepaper), we will be presenting preclinical data that demonstrate encouraging activity with SGN-LIV1A, an ADC in phase 1 clinical development for breast cancer, and the preclinical development of a novel ADC for multiple myeloma, SGN-CD352A, which uses our newest ADC technology. We will also highlight the progress we have made in the important field of immuno-oncology, demonstrating that auristatin-based ADCs may be particularly well suited for combination with checkpoint inhibitors and presenting exciting preclinical data for SEA-CD40 and a small molecule, 2-fluorofucose."

Multiple presentations are being featured at AACR (Free AACR Whitepaper) that highlight advances with Seattle Genetics’ proprietary ADC and empowered antibody pipeline and research programs. Abstracts can be found at www.aacr.org and include the following:

Preclinical data from a novel ADC candidate SGN-CD352A for multiple myeloma will be highlighted in a poster presentation on Monday, April 18, 2016 (Abstract #1195). SGN-CD352A utilizes Seattle Genetics’ newest ADC technology and is composed of an anti-CD352 antibody attached to a highly potent cytotoxic DNA-crosslinking agent called a pyrrolobenzodiazepine (PBD) dimer via a proprietary site-specific conjugation technology to a monoclonal antibody with engineered cysteines (EC-mAb).
SGN-LIV1A will be highlighted in a poster presentation on Tuesday, April 19, 2016 (Abstract #2966) focusing on the potential of combination use with standard of care chemotherapeutic agents, including doxorubicin, paclitaxel (Abraxane), carboplatin and various kinase inhibitors, for the treatment of breast cancer. SGN-LIV1A is in a phase 1 trial for the treatment of LIV-1-expressing metastatic breast cancer.
A preclinical analysis will highlight the novel small molecule, 2-fluorofucose (2FF) in a poster presentation on Tuesday, April 19, 2016 (Abstract #4005). 2FF blocks cellular incorporation of a sugar (fucose) and has been shown in preclinical studies to stimulate T-cell receptor signaling leading to enhanced activation of dendritic cells.
The path of SEA-CD40 from research to clinical development will be highlighted in a poster presentation on Wednesday, April 20, 2016 (Abstract #4994). SEA-CD40 is in an ongoing phase 1 trial for patients with advanced malignancies. SEA-CD40 is a novel immuno-oncology agent targeted to CD40 utilizing Seattle Genetics’ proprietary sugar-engineered antibody (SEA) technology to produce a non-fucosylated antibody.
Preclinical data evaluating brentuximab vedotin (ADCETRIS) immune activity will be presented in a poster presentation on Wednesday, April 20, 2016 (Abstract #4914). The preclinical data being presented demonstrate that brentuximab vedotin treated tumor cells may initiate an antitumor immune response and supports combination strategies with immuno-oncology regimens, such as the ongoing phase 1/2 clinical trials evaluating ADCETRIS and nivolumab (Opdivo) in relapsed Hodgkin and non-Hodgkin lymphoma.
About Seattle Genetics

OncoSec and Heat Biologics Present Data at the American Conference for Cancer Research (AACR) Annual Meeting

On April 18, 2016 OncoSec Medical Incorporated ("OncoSec") (NASDAQ: ONCS), a company developing DNA-based intratumoral cancer immunotherapies, reported that preclinical data from its collaboration with Heat Biologics, Inc (Press release, OncoSec Medical, APR 18, 2016, View Source [SID:1234510979]). ("Heat") focused on evaluating the combination of immunotherapy platforms were presented yesterday at the American Conference for Cancer Research (AACR) (Free AACR Whitepaper) Annual Meeting (www.aacr.org).

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!

In the poster entitled "In vivo intra-tumoral electroporation of gp96-Ig/Fc-OX40L stimulates CD8+ T cell cross-priming to tumor specific neoantigens" (Abstract #567), researchers concluded that combining Heat’s ComPACT vaccine with OncoSec’s intratumoral DNA electroporation delivery platform stimulated an expansion of neoantigen-specific CD8+ T cells, leading to a regression in both treated and untreated cancer lesions in two mouse studies (melanoma and colorectal cancer). OncoSec and Heat announced their collaboration last year to evaluate the preclinical efficacy of delivering Heat’s immunotherapy vaccines via OncoSec’s ImmunoPulse platform.

"We are excited by our collaboration with Heat and by these initial findings. The ability of the tumor’s microenvironment to evade immune recognition and remain non-immunogenic is a significant challenge, which we believe needs to be addressed when designing new immuno-therapies," said Robert H. Pierce, MD, OncoSec’s Chief Scientific Officer. "The initial feasibility data between our two platforms are encouraging and we are currently exploring the potential synergy between our platforms with both ComPACT and interleukin-12 expressing plasmids."

"In this first preclinical study demonstrating the feasibility of electroporating ComPACT DNA plasmids, we saw robust neoantigen T cell response and tumor regression in both treated and untreated tumors, indicating a systemic anti-tumor response that could be reflective of what we might see in metastatic lesions," said Taylor Schreiber, MD, PhD, Heat’s Chief Scientific Officer. "We believe that this approach is promising and that further studies are merited, especially as this combination approach has the potential to stimulate shared and private tumor antigens without introducing the complexities associated with personalized therapies."

Copies of the abstract are available and can be viewed online through the AACR (Free AACR Whitepaper) website at www.aacr.org. The poster is available in the Publications section of OncoSec’s website.

6-K – Report of foreign issuer [Rules 13a-16 and 15d-16]

On April 18, 2016 Oncolytics Biotech Inc. ("Oncolytics" or the "Company") (TSX: ONC) (OTCQX: ONCYF) (FRA: ONY) reported that two poster presentations covering preclinical work in multiple myeloma and colorectal cancer are being made by the Company’s research collaborators at the 2016 American Association of Cancer Research annual meeting being held from April 16th to 20th, 2016 in New Orleans, LA (Filing, 6-K, Oncolytics Biotech, APR 18, 2016, View Source [SID:1234510978]).

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!

"Both multiple myeloma and colorectal cancer are target indications for later-stage clinical testing of REOLYSIN," said Dr. Brad Thompson, President and CEO of Oncolytics. "These important preclinical findings continue to advance our understanding of how REOLYSIN interacts with the immune system and how immunomodulatory interventions could improve its cancer cell killing capabilities."

The first abstract/poster is titled "Successful oncolytic virotherapy in a bortezomib resistant syngeneic mouse model of multiple myeloma: implications for translation significance," and was authored by Thirukkuamaran, et al. Using the VK*MYC bortezomib resistant transplantable multiple myeloma mouse model, the authors demonstrated that mice harboring bortezomib insensitive multiple myeloma tumors significantly responded to reovirus treatment. These data are supportive of previous and ongoing preclinical and clinical work in this indication and the Company is currently enrolling patients in a Phase 1b study of REOLYSIN in combination with bortezomib in patients with relapsed or refractory multiple myeloma.

The second abstract/poster is titled "Toll like receptor 3 as an immunotherapeutic target for Kras mutated colorectal cancer," and was authored by Goel, et al. The authors hypothesized that effective expression of toll like receptors 3 ("TLR3") would dampen the infection potential of reovirus through the mounting of an innate immune response. Using a xenograft model with HCT116 colorectal cancer cells, those with TLR3 downregulated cells showed improved control of tumor growth with reovirus treatment compared to those expressing TLR3 (p=0.04). Down regulation of the host immune response improved virus mediated cell cytotoxicity and the findings could result in improved and beneficial killing of cancer cells by reovirus.

Preclinical Data Presented at AACR Demonstrate that Combining NKTR-214 with Checkpoint Blockade is Superior to Dual Checkpoint Inhibition in Increasing Clonality of the T Cell Receptor (TCR) Repertoire and T Cell Tumor Infiltration

On April 18, 2016 Nektar Therapeutics (NASDAQ: NKTR) reported new preclinical data for the company’s investigational immuno-stimulatory cytokine therapy, NKTR-214, which demonstrate both its activity as a single-agent and its synergistic activity with checkpoint blockade (Press release, Nektar Therapeutics, APR 18, 2016, View Source [SID:1234510977]). In a TCR repertoire analysis conducted to assess both anti-tumor T cell clonality and T cell tumor infiltration (TIL clonality and infiltration), the combination of NKTR-214 and a checkpoint inhibitor resulted in dramatically higher increases for both parameters as compared to dual checkpoint inhibition. These data were presented at the American Association for Cancer Research (AACR) (Free AACR Whitepaper) Annual Meeting in New Orleans, LA on April 17, 2016.

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!

TIL clonality establishes the frequency of T cells with a specific TCR Vβ and TCR Jβ chain usage at the tumor site, which suggests increased CD8-positive effector T cell function against the specific tumor.1 The concomitant presence of both TIL clonality and infiltration has been significantly correlated with clinical response and better survival outcomes in patients.1 In a CT26 colon carcinoma model, sequencing for TCRs in the tumor was conducted using Adaptive Biotechnologies’ ImmunoSEQ platform. Measurements of TIL clonality and infiltration were assessed seven days after treatment. NKTR-214 as a single-agent led to superior increases in TIL clonality and infiltration as compared to either anti-CTLA-4 or anti-PD-1 therapy alone. The combination of NKTR-214 with either mode of checkpoint inhibition led to superior increases in both TIL clonality and infiltration relative to the combination of CTLA-4 and PD-1 inhibitors. The highest increases occurred when NKTR-214 was added to an anti-PD-1 therapy.

"We are particularly excited by these new preclinical data for the combination of NKTR-214 with a checkpoint inhibitor which show an induced oligoclonal T cell response along with high TIL infiltration, "said Dr. Jonathan Zalevsky, Vice President of Biology at Nektar Therapeutics. "These data demonstrate that the addition of NKTR-214 to either an anti-CTLA-4 or anti-PD-1 agent could improve T cell clonality differences over checkpoint blockade therapies."

NKTR-214 is an investigational CD122-biased agonist currently in Phase 1/2 clinical development. NKTR-214 is designed to stimulate the patient’s own immune system to kill tumor cells by preferentially activating production of specific immune cells which promote tumor killing, including CD8-positive T cells and Natural Killer (NK) cells, within the tumor micro-environment. CD122, which is also known as the Interleukin-2 receptor beta subunit, is a key signaling receptor that is known to increase proliferation of these types of T cells.2

In studies conducted in multiple established tumor models presented at AACR (Free AACR Whitepaper), NKTR-214 demonstrated activity as both a single-agent and when co-dosed with either an anti-PD-1 agent or an anti-CTLA-4 agent. In a tumor re-challenge study conducted in an EMT6 colon carcinoma model, sequential dosing of anti-CTLA-4 followed by NKTR-214 resulted in durable immunity, with complete responders resisting a tumor re-challenge. Following a second re-challenge, 100% of the animals remained tumor-free without additional treatment.

"Our latest preclinical findings continue to show that treatment with NKTR-214 either as a single-agent or in combination with checkpoint blockade is superior to single or dual checkpoint inhibition in multiple models," added Steve Doberstein, PhD, Senior Vice President and Chief Scientific Officer of Nektar Therapeutics. "These new data emphasize the cellular mechanisms underlying the remarkable efficacy of NKTR-214 in our preclinical models of cancer."

The data presentation at AACR (Free AACR Whitepaper) entitled, "Durable antitumor activity of the CD122-biased immuno-stimulatory cytokine NKTR-214 combined with immune checkpoint blockade," can be accessed at View Source

In preclinical studies, NKTR-214 demonstrated a highly favorable mean ratio of 450:1 within the tumor micro-environment of CD8-positive effector T cells relative to regulatory T cells.3 Furthermore, the pro-drug design of NKTR-214 enables an antibody-like dosing regimen for an immuno-stimulatory cytokine.4

About the NKTR-214 Phase 1/2 Clinical Study

A Phase 1/2 clinical study is underway to evaluate NKTR-214 in patients with advanced solid tumors, including melanoma, renal cell carcinoma and non-small cell lung cancer. The first stage of this study, which is expected to be complete in the second half of 2016, is evaluating escalating doses of single-agent NKTR-214 treatment in approximately 20 patients with solid tumors. The primary objective of the first stage of the study is to evaluate the safety and efficacy of NKTR-214 and to identify a recommended Phase 2 dose. In addition, the study will also assess the immunologic effect of NKTR-214 on tumor-infiltrating lymphocytes (TILs) and other immune cells in both blood and tumor tissue, and it will also include TCR repertoire profiling. Dose expansion cohorts are planned to evaluate NKTR-214 in specific tumor types, including melanoma, renal cell carcinoma and non-small cell lung cancer.

The NKTR-214 clinical study is being conducted initially at two primary investigator sites: MD Anderson Cancer Center under Drs. Patrick Hwu and Adi Diab; and Yale Cancer Center, under Drs. Mario Sznol and Michael Hurwitz. Patients and physicians interested in the ongoing NKTR-214 Study can visit the "Clinical Trials" section of www.mdanderson.org using identifier 2015-0573 or visit View Source

IPI-549 Alters the Immune-Suppressive Microenvironment and Enhances the Activity of Checkpoint Inhibitors in Preclinical Models

On April 18, 2016 Infinity Pharmaceuticals, Inc. (NASDAQ: INFI) reported new preclinical data for IPI-549, an orally administered immuno-oncology development candidate that selectively inhibits phosphoinositide-3-kinase gamma (PI3K-gamma) (Press release, Infinity Pharmaceuticals, APR 18, 2016, View Source;p=RssLanding&cat=news&id=2157820 [SID:1234510975]). Preclinical data in multiple solid tumor models demonstrate that IPI-549 targets immune cells and alters the immune-suppressive microenvironment, promoting an anti-tumor immune response that leads to tumor growth inhibition. Data also demonstrate that IPI-549 enhances the effects of checkpoint inhibitors, resulting in improved survival in murine models. These data were presented at the American Association for Cancer Research (AACR) (Free AACR Whitepaper) Annual Meeting 2016 in New Orleans, Louisiana. A Phase 1 clinical study is under way to explore the safety and activity of IPI-549 both as a monotherapy and in combination with anti-PD-1 antibody therapy, a type of checkpoint inhibitor.

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!

"While advances in our understanding of the immune response to cancer have resulted in new therapies for patients, additional treatments are needed that can offer even more patients the chance for prolonged survival," stated Jedd Wolchok, M.D., Ph.D., chief of Melanoma and Immunotherapeutics Service, Lloyd J. Old/Ludwig Chair in Clinical Investigation Department of Medicine and Ludwig Center, at Memorial Sloan Kettering Cancer Center and lead investigator for the Phase 1 clinical study of IPI-549. "Emerging data from our collaboration with Infinity’s discovery team provide additional rationale for combining IPI-549 with checkpoint inhibitors, and I am pleased to be leading the Phase 1 study for this program."

Preclinical Data for IPI-549 Presented at the AACR (Free AACR Whitepaper) Annual Meeting 2016 (Abstract #554)
Infinity researchers, in collaboration with researchers at Memorial Sloan Kettering Cancer Center, presented preclinical data for IPI-549 in a poster entitled, "Checkpoint blockade therapy is improved by altering the immune suppressive microenvironment with IPI-549, a potent and selective inhibitor of PI3K-gamma, in preclinical models."

In preclinical models, treatment with IPI-549 leads to a decrease in tumor-associated immune suppressive myeloid cells. IPI-549 treatment also leads to a decrease in FOXP3 T-regulatory cells, which have immune-suppressive effects, and an increase in intratumoral CD8+ T-cells, which are known to play a role in inhibiting tumor growth. Taken together, these data suggest that through its effect on myeloid cells and T-cells, IPI-549 has the potential to disrupt the immune-suppressive microenvironment and enable a heightened anti-tumor immune response.

Preclinical data in murine models show that treatment with IPI-549 in combination with anti-CTLA4 or anti-PD-L1, two types of checkpoint inhibitors, results in greater tumor growth inhibition compared to monotherapy treatment. Additionally, IPI-549 in combination with anti-PD-1 increased the number of complete responses and improves survival. Re-implantation of tumor cell lines into mice that had achieved complete responses revealed low or no tumor engraftment, suggesting sustained tumor-specific immune protection.

These data provide additional preclinical rationale for the ongoing Phase 1 clinical study designed to explore the safety and activity of IPI-549 as a monotherapy and in combination with anti–PD-1 antibody therapy in patients with selected solid tumors, including non-small cell lung cancer and melanoma (ClinicalTrials.gov identifier NCT02637531).

Infinity is also developing duvelisib, an investigational, oral, dual inhibitor of PI3K-delta and PI3K-gamma. The PI3K pathway is also known to play a critical role in regulating the growth and survival of certain types of blood cancers. Duvelisib is being evaluated in registration-focused studies, including DYNAMOTM, a Phase 2 study in patients with refractory indolent non-Hodgkin lymphoma (iNHL), BRAVURA, a Phase 3 study in patients with relapsed iNHL, and DUOTM, a Phase 3 study in patients with relapsed/refractory chronic lymphocytic leukemia. For additional information about clinical studies of duvelisib, please visit www.infi.com or www.clinicaltrials.gov.

About IPI-549
IPI-549 is an orally administered immuno-oncology development candidate that selectively inhibits PI3K-gamma. In preclinical studies, IPI-549 inhibits immune-suppressive macrophages within the tumor microenvironment, whereas other immunotherapies such as checkpoint modulators more directly target immune effector cell function. As such, IPI-549 may have the potential to treat a broad range of solid tumors and represents a potentially complementary approach to restoring anti-tumor immunity in combination with other immunotherapies such as checkpoint inhibitors.

Duvelisib and IPI-549 are investigational compounds and their safety and efficacy have not been evaluated by the U.S. Food and Drug Administration or any other health authority.