TG Therapeutics, Inc. Announces Oral Data Presentation for TGR-1202 in Combination with Ibrutinib in Patients with Relapsed or Refractory CLL or MCL at the 58th American Society of Hematology Annual Meeting

On December 5, 2016 TG Therapeutics, Inc. (NASDAQ:TGTX), reported the presentation of combination data from a Phase 1b study evaluating TGR-1202, the Company’s once-daily PI3K delta inhibitor, in combination with ibrutinib, the oral Bruton’s tyrosine kinase (BTK) (Press release, TG Therapeutics, DEC 5, 2016, View Source [SID1234516937]). This study is being run in collaboration with the Blood Cancer Research Partnership (BCRP) and Dana-Farber Cancer Institute (DFCI), Boston, MA. Data from this trial were presented today by the Principal Investigator, Matthew S. Davids, MD, of Dana-Farber Cancer Institute, during an oral session at the 58th American Society of Hematology (ASH) (Free ASH Whitepaper) annual meeting in San Diego, CA.

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!

Michael S. Weiss, the Company’s Executive Chairman and Interim CEO commenting on the data said, "We and our investigators continue to be impressed with the ease of combining TGR-1202 with other novel agents. Here Dr. Davids demonstrated that the double blockade of the BCR pathway with dual oral once-daily inhibitors can be performed safely and conveniently, and derives high response rates in patients with both CLL and MCL. This all oral approach offers a unique and highly active alternative for patients who do not want to receive infused therapies but still want to advance their treatment beyond single agent ibrutinib, which may offer multiple benefits over single agent therapy. With one bone marrow confirmed complete response (CR) and 5 additional deep responses nearing radiographic CR out of the 17 evaluable CLL patients, we and our investigators believe we are seeing activity beyond what one might expect from either of these agents alone. We want to thank Dr. Davids and his collaborators at DFCI and the Leukemia & Lymphoma Society for providing support for this important investigator driven research and we look forward to follow-up data in the future from this study and data from our own triple therapy of TG-1101 plus TGR-1202 plus ibrutinib, which we are targeting for presentation next year."

The following summarizes the key highlights from this oral presentation which occurred today:

Oral Presentation: TGR-1202 in Combination with Ibrutinib in Patients with Relapsed or Refractory CLL or MCL: Preliminary Results of a Multicenter Phase I/Ib Study (Abstract Number 641)

This oral presentation includes data from patients with relapsed or refractory chronic lymphocytic leukemia (CLL) or mantle cell lymphoma (MCL) treated with TGR-1202 in combination with Ibrutinib. 31 patients were evaluable for safety (18 CLL patients and 13 MCL patients), of which 28 patients were available for efficacy (17 CLL patients and 11 MCL patients.) CLL patients had a median of 1.5 prior lines of therapy (range 1-6), with 2 patients receiving prior ibrutinib and 4 receiving prior PI3K inhibitors. MCL patients had a median of 3 prior lines of therapy (range 2-5), with 2 patients also receiving prior ibrutinib.

Highlights from this oral presentation include:

88% (15 of 17) Overall Response Rate (ORR) (including Complete Response (CR), Partial Response (PR), and Partial Response with lymphocytosis (PR-L)) in patients with CLL, with 1 patient achieving a bone marrow confirmed CR and 5 patients with a > 80% nodal reduction, nearing radiographic CR
1 year progression free survival (PFS) and overall survival (OS) for CLL is 94% (n=17), with the longest patient on study approaching two years
73% (8/11) ORR in patients with MCL, with clinical benefit observed in two additional patients
1 year PFS and OS for MCL is 37% and 52%, respectively (n=11)
The combination appears well tolerated across all patients with no grade 3/4 transaminitis (liver toxicity), diarrhea, colitis or pneumonitis observed
PRESENTATION DETAILS:

The above referenced presentation is available on the Company’s website at www.tgtherapeutics.com, located on the Publications page.

Preclinical Data on IMGN632, a Novel CD123-Targeting ADC, Presented at ASH Annual Meeting

On December 5, 2016 ImmunoGen, Inc. (Nasdaq: IMGN), a leader in the expanding field of antibody-drug conjugates (ADCs) for the treatment of cancer, reported that preclinical data on IMGN632, a novel CD123-targeting ADC, were presented today at the 58th American Society of Hematology (ASH) (Free ASH Whitepaper) Annual Meeting in San Diego, CA (Press release, ImmunoGen, DEC 5, 2016, View Source [SID1234516935]).

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!

CD123 is an attractive target due to its elevated expression in acute myeloid leukemia (AML). IMGN632 uses ImmunoGen’s new family of indolino-benzodiazepine cancer-killing agents, called IGNs. ImmunoGen designed IGNs to be highly potent and to alkylate DNA without crosslinking it. Specifically, IMGN632 uses the Company’s DGN549 payload and incorporates novel linker and conjugation technology.

"We developed our DNA-alkylating IGN payloads to meet the dual challenges of achieving high potency against target cells, while having a tolerability profile that enables continued patient treatment," said Richard Gregory, Ph.D., Executive Vice President and Chief Scientific Officer of ImmunoGen. "These preclinical data demonstrate that IMGN632 has the potential for broad and potent activity in patients with AML and an improved tolerability profile."

The data presented at ASH (Free ASH Whitepaper) (oral abstract #768) compared IMGN632, an ADC with an alkylating IGN, to a version of IMGN632 with a crosslinking payload. In vitro cytotoxic activity was compared in multiple AML cell lines. Both ADCs were found to be highly active against AML cells, including those with poor prognostic markers (FLT3-ITD, P53, MDR1), and were approximately 100-fold more active on AML patient samples than gemtuzumab ozogamicin.

Both ADCs exhibited similar efficacy in human AML xenograft models; however, the effects of the ADCs in toxicity studies were very different. While IMGN632, the alkylating ADC, was well tolerated at the dose tested, the crosslinking ADC showed persistent delayed toxicity (weight loss) at less than half the dose.

In addition, on normal bone marrow cells, IMGN632 was approximately 50-fold less toxic than the crosslinking ADC, while retaining high potency against AML cells.

These results show that IMGN632 has potent selective activity against AML cells with lower cytotoxicity to normal myeloid progenitor cells than an ADC designed to crosslink DNA activity. These data suggest IMGN632 has the potential to be a highly potent yet tolerable ADC for AML patients.

Supporting preclinical data were also presented at ASH (Free ASH Whitepaper) in which IMGN632 showed compelling activity in AML xenograft models (abstract #2832).

The Company plans to submit an IND application and initiate clinical testing of IMGN632 in 2017.

Preclinical data were also presented at ASH (Free ASH Whitepaper) on IMGN779, a potent CD33-targeting ADC using an IGN payload, (abstract #1645) from a combination study of IMGN779 with a PARP inhibitor (olaparib). The data demonstrated enhanced activity in several AML models including patient derived tumor cells and a disseminated AML xenograft model. IMGN779 is currently being evaluated in a Phase 1 study as a monotherapy in AML.

About Acute Myeloid Leukemia
Acute myeloid leukemia (AML) is a cancer of the bone marrow cells that produce white blood cells. It causes the marrow to increasingly generate abnormal immature white blood cells (blasts) that do not mature into effective infection-fighting cells. The blasts quickly fill the bone marrow, impacting the production of normal platelets and red blood cells. The resulting deficiencies in normal blood cells leave the patient vulnerable to infections, bleeding problems and anemia.

In 2016, it is estimated that nearly 20,000 new cases of AML will be diagnosed in the U.S. and more than 10,000 people will die from the disease.1

Bellicum Pharmaceuticals Announces GoCAR-T and GoTCR Preclinical Presentations at the American Society of Hematology 2016 Annual Meeting

On December 5, 2016 Bellicum Pharmaceuticals, Inc. (Nasdaq: BLCM), a clinical stage biopharmaceutical company focused on discovering and developing novel cellular immunotherapies for cancers and orphan inherited blood disorders, reported the presentation of preclinical results on the Company’s GoCAR-T and GoTCR technologies at the 58th American Society of Hematology (ASH) (Free ASH Whitepaper) Annual Meeting in San Diego, California (Press release, Bellicum Pharmaceuticals, DEC 5, 2016, View Source;p=RssLanding&cat=news&id=2227609 [SID1234516934]).

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 pleased to report further supportive preclinical data on the utility of our proprietary iMC activation switch, which is incorporated into our BPX-601 GoCAR-T product candidate now entering a Phase 1 clinical study," commented Tom Farrell, President and CEO of Bellicum Pharmaceuticals. "We believe our novel technology provides a powerful and unique solution for overcoming the efficacy and safety challenges of T-cell therapies, especially when targeting solid tumors."

The Company’s GoCAR-T platform incorporates an inducible MyD88/CD40 (iMC) costimulatory switch, which requires presence of both a target antigen and rimiducid to trigger the full effect of CAR T cell activity. The presence of rimiducid and antigen results in upregulation of cytokines such as IL2, leading to T-cell proliferation, persistence and improved anti-tumor efficacy. Unlike traditional CAR T constructs, GoCAR-T is designed to support persistence of CAR T cells in the body in the absence of cancer antigen to provide continued anti-tumor surveillance.

In a poster presentation titled, "Inducible MyD88/CD40 (iMC) Costimulation Provides Ligand-Dependent Tumor Eradication By CD123-Specific Chimeric Antigen Receptor T Cells," Bellicum scientists targeted CD123, which is highly expressed in acute myeloid leukemia (AML) and leukemic stem cells. Results demonstrated that GoCAR-T removed CD123-positive leukemic cells in animal models through rimiducid-activated costimulation. Conversely, infrequent costimulation with rimiducid led to reduced activity of CAR T cells, supporting the technology’s potential to provide control over the activation, expansion and persistence of cells to achieve a desired level of safety and anti-tumor potency.

Additional data were presented on the Company’s GoTCR technology, which also uses an iMC costimulatory switch. The presence of GoTCR and rimiducid triggers the release of cytokines that upregulate MHC (major histocompatibility complex) on tumor cells, exposing them to potent immune response by both engineered and endogenous T cells. In the study outlined in a poster presentation titled, "Inducible MyD88/CD40 (iMC) Enhances Proliferation and Survival of Tumor-Specific TCR-Modified T Cells and Improves Anti-Tumor Efficacy in Myeloma," T cells were engineered to express tumor antigen-specific T-cell receptors (TCRs) targeting preferentially-expressed antigen in melanoma (PRAME) or Bob1. PRAME is overexpressed in a wide variety of cancers including melanoma, sarcoma and several types of leukemias. Bob1 is also found to be highly expressed in certain leukemias, along with lymphomas and myelomas. Results demonstrated that the rimiducid-driven iMC costimulatory switch provided potent T-cell activation, proliferation and persistence, synergizing with signals from PRAME- or Bob1-targeted TCRs for improved anti-tumor efficacy in vitro and in vivo.

TG Therapeutics, Inc. Announces Preclinical Data Presentations for TGR-1202 at the 58th American Society of Hematology Annual Meeting

On December 5, 2016 TG Therapeutics, Inc. (NASDAQ:TGTX), reported the presentation of two preclinical data sets, one oral presentation and one poster presentation, for TGR-1202, the Company’s once-daily PI3K delta inhibitor, at the 58th American Society of Hematology (ASH) (Free ASH Whitepaper) annual meeting in San Diego, California (Press release, TG Therapeutics, DEC 5, 2016, View Source [SID1234516932]).

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!

Michael S. Weiss, the Company’s Executive Chairman and Interim Chief Executive Officer, stated, "We want to thank the teams at Columbia and Moffitt for their extensive laboratory work on TGR-1202 to better understand the mechanism of action and impact on the immune system. The preclinical data they have generated helps to better explain and perhaps offer a rationale for the differentiated safety profile seen with TGR-1202 as compared to earlier generation PI3K delta inhibitors. We believe these preclinical findings along with the robust safety and efficacy data we have observed in the clinic, support our belief that TGR-1202 is a differentiated best in class PI3K delta inhibitor. We look forward to continuing our research collaborations with Columbia and Moffitt and to presenting updated safety and efficacy data for TGR-1202 to further confirm its unique profile."

"Dr. Deng’s presentation today has really begun to shed some long-needed light on the important differences among the PI3K delta inhibitors. His work has identified that a novel kinase important in the PI3K pathway, CK-1epsilon, is uniquely inhibited by TGR-1202, which may explain the drug’s effects on c-Myc. These chemical differences may also help to explain the important immunologic differences in the safety profiles of these agents," stated Dr. Owen A. O’Connor, Professor of Medicine and Experimental Therapeutics, Director Lymphoid Malignancies at Columbia Presbyterian Medical Center.

The following summarizes the oral presentation and poster presentation which occurred yesterday:

Oral Presentation: Silencing c-Myc Translation as a Therapeutic Strategy through Targeting PI3K Delta and CK1 Epsilon in Hematological Malignancies (Abstract Number 291)

This oral presentation includes data from the manuscript titled, "Silencing c-Myc Translation as a Therapeutic Strategy through Targeting PI3K Delta and CK1 Epsilon in Hematological Malignancies," which was recently published in Blood, the Journal of the American Society of Hematology (ASH) (Free ASH Whitepaper). The presentation was delivered by Changchung Deng, MD, PhD of Columbia Presbyterian Medical Center and included the following highlights:

TGR-1202 and carfilzomib, but not combinations of other drugs in the same classes, synergistically inhibit c-Myc translation and c-Myc dependent gene transcription, by potently inhibiting phosphorylation of 4E-BP1;
TGR-1202 and carfilzomib synergistically induce apoptosis in lymphoma cells through targeting c-Myc, whereas the other combinations did not;
TGR-1202, but not idelalisib or duvelisib, was found to uniquely inhibit casein kinase-1 (CK1) epsilon; and
Based on this extensive preclinical work, the Company recently announced the launch of a Phase 1/2 study to evaluate the safety and efficacy of TGR-1202 in combination with carfilzomib, in patients with relapsed or refractory lymphoma.
Poster Presentation: Modulation of T Cell Compartment in a Preclinical CLL Murine Model By a Selective PI3K Delta Inhibitor, TGR-1202 (Abstract Number 3236)

This poster presentation included preclinical data describing the differential regulation of human T-cells by TGR-1202 in a preclinical CLL murine model. Highlights from this poster include:

Both TGR-1202 and duvelisib oral administration demonstrated comparable efficacy by reducing CLL burden over time in leukemic mice;
TGR-1202 and duvelisib both targeted the T cell population in vivo, however:
TGR-1202 relatively maintained the number of Tregs and Th17 cells and expression of functional markers on Tregs compared to duvelisib treatment in vivo and ex vivo; and
Duvelisib resulted in greater disruption of Treg/Th17 ratio compared to TGR-1202 in vivo, which may have implications for occurrence of autoimmune-like organ toxicity.
PRESENTATION DETAILS:
Copies of the above referenced presentations are available on the Company’s website at www.tgtherapeutics.com, located on the Publications page.

TG THERAPEUTICS INVESTOR & ANALYST EVENT DETAILS:
TG Therapeutics will also host an investor and analyst reception on Monday, December 5th, 2016 beginning at 8:00pm PT. The event will take place at the Marriott Gaslamp, in San Diego, California, in the Presidio AB Ballroom.
NOTE: This event will be webcast live and will be available on the Events page, located within the Investors & Media section of the Company’s website at www.tgtherapeutics.com, as well as archived for future review. This event will also be broadcast via conference call. In order to access the conference line, please call 1-877-407-8029 (U.S.), 1-201-689-8029 (outside the U.S.), and reference Conference Title: TG Therapeutics 2016 Investor & Analyst Event.

Seattle Genetics Highlights Phase 1 Vadastuximab Talirine (SGN-CD33A; 33A) Data Presentations, Including Combination Therapy with HMAs, in Patients with Acute Myeloid Leukemia at ASH Annual Meeting

On December 5, 2016 Seattle Genetics, Inc. (Nasdaq: SGEN), a global biotechnology company, reported three oral data presentations on vadastuximab talirine (SGN-CD33A; 33A) in patients with acute myeloid leukemia (AML) at the 58th American Society of Hematology (ASH) (Free ASH Whitepaper) Annual Meeting and Exposition taking place in San Diego, California, December 3-6, 2016 (Press release, Seattle Genetics, DEC 5, 2016, View Source;p=RssLanding&cat=news&id=2227547 [SID1234516931]). The data included updated results from an ongoing phase 1 clinical trial evaluating 33A in combination with hypomethylating agents (HMAs; azacitidine, decitabine) in frontline older AML patients. Further oral presentations focused on results from phase 1 clinical trials evaluating 33A in three distinct settings, including 1) as monotherapy in newly diagnosed older AML patients, 2) in combination with high-dose cytarabine for younger AML patients in first remission and 3) as monotherapy maintenance for younger AML patients who have completed frontline therapy or after allogeneic stem cell transplant. 33A is an investigational antibody-drug conjugate (ADC) targeted to CD33, a protein which is expressed on leukemic cells in nearly all AML patients.

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!

Seattle Genetics is broadly evaluating 33A across multiple lines of therapy in patients with myeloid malignancies. In addition to the clinical trials presented at ASH (Free ASH Whitepaper) this year, 33A is currently being evaluated in combination with HMAs in the ongoing global phase 3 CASCADE study. This trial is randomized, double-blind, and being conducted at multiple centers globally to evaluate if 33A in combination with azacitidine or decitabine can extend overall survival compared to azacitidine or decitabine alone in approximately 500 older patients with newly diagnosed AML. Additional phase 1 and 2 clinical trials for frontline younger AML and frontline myelodysplastic syndrome (MDS) are also underway. More information about 33A and ongoing clinical trials can be found at www.ADC-CD33.com.

"We are pleased with the growing body of data demonstrating that vadastuximab talirine, also known as 33A, has a promising overall tolerability and activity profile in clinical trials for patients with AML," said Jonathan Drachman, M.D., Chief Medical Officer and Executive Vice President, Research and Development at Seattle Genetics. "We are committed to improving the therapeutic options for AML patients through innovative, targeted approaches. Our most advanced 33A clinical study, CASCADE, is a randomized phase 3 trial designed to test 33A in combination with hypomethylating agents, or HMAs, in approximately 500 older patients with newly diagnosed AML. Based on the encouraging data presented at ASH (Free ASH Whitepaper), we believe 33A has the potential for clinical development in multiple AML settings, with the goal of providing new treatment options for patients struggling with this aggressive and life-threatening disease."

"AML therapy has not meaningfully changed over the past 40 years, and there is a dire need for improved treatment options. Older AML patients have a particularly poor prognosis with the standard of care, hypomethylating agents or HMAs," said Amir Fathi, M.D., Massachusetts General Hospital Cancer Center. "I am pleased with the balance of activity and tolerability we have observed in phase 1 clinical trials evaluating 33A both as monotherapy and combination therapy in AML patients. For older patients with newly diagnosed AML, the 73 percent remission rate of 33A in combination with HMAs, with 50 percent of those patients negative for minimal residual disease, signals promise in improving long-term outcomes."

The following data from the ongoing phase 1 studies evaluating 33A combination therapy or monotherapy in AML patients were presented:

Vadastuximab Talirine Plus Hypomethylating Agents: A Well-Tolerated Regimen with High Remission Rate in Frontline Older Patients With Acute Myeloid Leukemia (Abstract #591, oral presentation on Monday, December 5, 2016 at 7:30 a.m. PT)

Outcomes for AML patients who are older or ineligible to receive standard chemotherapy remain poor. HMAs are frequently used in this setting, but efficacy is limited. Updated results from an ongoing phase 1 study evaluating 33A in combination with HMAs (either azacitidine or decitabine) in newly diagnosed older AML patients were presented by Dr. Amir Fathi, Massachusetts General Hospital Cancer Center.

Data were reported from 53 frontline AML patients with a median age of 75 years and predominantly intermediate or adverse cytogenetic risk who had declined intensive therapy. Regarding additional poor-prognosis indicators, 42 percent of patients had evidence of underlying myelodysplasia, 11 percent had FLT3-mutated disease and 43 percent had secondary AML, which is AML that arises from prior chemotherapy, a pre-existing MDS or myeloproliferative disease. Key findings include:

Of 49 patients evaluable for response, complete remission (CR) or CR with incomplete platelet or neutrophil recovery (CRi) was observed in 36 patients (73 percent).
Remissions were observed in higher-risk patients, including 17 of 22 patients (77 percent) with secondary AML, five of five patients (100 percent) who were FLT3/ITD positive and 17 of 26 patients (65 percent) age 75 or older.
Eighteen of the 36 patients (50 percent) who achieved remission (CR or CRi) were negative for minimal residual disease (MRD), which means no cancer could be detected with a sensitive test.
With a median follow-up of 14.7 months, median overall survival for all patients was 11.3 months and 28 percent of patients remained alive and on study as of last follow-up. The 30- and 60-day mortality rates were two and eight percent, with no treatment-related deaths occurring during that time.
For patients who achieved MRD-negative remission, the median survival had not yet been reached.
The most common Grade 3 or 4 treatment-emergent adverse events occurring in 20 percent or more of patients were thrombocytopenia, febrile neutropenia, anemia and neutropenia.
The most common Grade 1 and 2 treatment-emergent adverse events occurring in 20 percent or more of patients were fatigue, nausea, constipation, peripheral edema and decreased appetite.
Vadastuximab Talirine Monotherapy in Older Patients with Treatment Naive CD33-Positive Acute Myeloid Leukemia (Abstract #590, oral presentation on Monday, December 5, 2016 at 7:15 a.m. PT)

Interim results from 93 patients in the ongoing phase 1 study evaluating 33A monotherapy in AML patients were previously presented at the 2015 ASH (Free ASH Whitepaper) Annual Meeting. New results describing the safety and activity of the recommended 33A monotherapy dose of 40 micrograms per kilogram (mcg/kg) in an expansion cohort of treatment-naïve older AML patients were presented by Dr. Anjali Advani, Cleveland Clinic.

Data were reported from 27 treatment-naïve older AML patients with a median age of 74 years and intermediate or adverse cytogenetic risk of 70 percent and 26 percent, respectively. Regarding additional poor-prognosis indicators, 48 percent of patients had evidence of underlying myelodysplasia and 22 percent had FLT3 mutated disease. Key findings include:

Of the 26 patients evaluable for response, remission (CR or CRi) was observed in 15 patients (58 percent), with a median time to remission of 1.4 months.
Forty-three percent of patients who achieved remission were MRD negative.
Responses were observed in higher-risk patients, with remissions achieved in seven of 12 patients (58 percent) with underlying myelodysplasia and three of four patients (75 percent) who were positive for FLT3/ITD.
The 30- and 60-day mortality rates were zero and 15 percent, respectively. The median overall survival for all patients was seven months.
The most common Grade 3 or higher treatment-emergent adverse events occurring in 20 percent or more of patients were thrombocytopenia, febrile neutropenia and anemia.
The most common Grade 1 and 2 treatment-emergent adverse events occurring in 20 percent or more of patients were peripheral edema, decreased appetite, fatigue, diarrhea and dizziness.
A Phase 1b Study of Vadastuximab Talirine as Maintenance and in Combination with Standard Consolidation for Patients with Acute Myeloid Leukemia (Abstract #340, oral presentation on Sunday, December 4, 2016 at 10:15 a.m. PT)

Interim results from an ongoing phase 1b study evaluating 33A in the AML post-remission setting, as a single agent for maintenance therapy or in combination with consolidation therapy (high-dose cytarabine; HiDAC), were presented by Dr. Jay Yang, Karmanos Cancer Institute.

About Acute Myeloid Leukemia

Acute myeloid leukemia, also called acute myelocytic leukemia or AML, is an aggressive cancer of the bone marrow and blood that progresses rapidly without treatment. Cancerous cells called leukemic blasts multiply and crowd out normal cells in the bone marrow and interfere with normal blood cell production leading to anemia, infection, and bleeding. According to the SEER database and Kantar Health Sciences, in 2016 approximately 33,000 new cases of AML (mostly in adults) will be diagnosed in the U.S. and Europe. In the U.S. alone, nearly 10,500 deaths will occur from AML this year. Treatment options for AML have remained virtually unchanged for nearly 40 years and frontline treatment consists primarily of chemotherapy. A subset of patients (typically those over 60 years of age) cannot tolerate such therapy and are typically given lower intensity therapies agents, supportive care, or are recommended for clinical trials.

About Vadastuximab Talirine (SGN-CD33A)

Vadastuximab talirine (SGN-CD33A; 33A) is a novel investigational ADC targeted to CD33 utilizing Seattle Genetics’ proprietary ADC technology. CD33 is expressed on most AML and MDS blast cells. The CD33 engineered cysteine antibody is stably linked to a highly potent DNA binding agent called a pyrrolobenzodiazepine (PBD) dimer via site-specific conjugation technology (EC-mAb). PBD dimers are significantly more potent than systemic chemotherapeutic drugs and the EC-mAb technology allows uniform drug-loading onto an ADC. The ADC is designed to be stable in the bloodstream and to release its potent cell-killing PBD agent upon internalization into CD33-expressing cells.

33A was granted Orphan Drug Designation by both the U.S. Food and Drug Administration (FDA) and the European Commission for the treatment of AML. FDA orphan drug designation is intended to encourage companies to develop therapies for the treatment of diseases that affect fewer than 200,000 individuals in the United States.