Amgen To Present At the Piper Jaffray And Evercore ISI Healthcare Conferences

On November 21, 2018 Amgen (NASDAQ: AMGN) reported that it will present at the Piper Jaffray Healthcare Conference at 8:30 a.m. ET on Tuesday, Nov. 27, 2018, in New York City and at the Evercore ISI Healthcare Conference at 11 a.m. ET on Wednesday, Nov. 28, 2018, in Boston (Press release, Amgen, NOV 21, 2018, View Source [SID1234531552]). Elliott M. Levy, M.D., senior vice president of Global Development at Amgen, will present at the conferences. Live audio of each presentation can be accessed from the Events Calendar on Amgen’s website, www.amgen.com, under Investors. A replay of each webcast will also be available on Amgen’s website for at least 90 days following the event.

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!


Cellanyx Publishes Clinical Proof-of-Concept Study in Urology of First-in-Class Live Tumor Cell, Phenotypic Biomarker Test to Risk Stratify Prostate Cancer Patients

On November 21, 2018 Cellanyx and clinical collaborators reported results of a prostate cancer clinical study demonstrating the ability of the company’s live tumor cell phenotypic biomarker test to identify patients with low and intermediate grade prostate cancer at risk of aggressive disease (Press release, Cellanyx, NOV 21, 2018, View Source [SID1234531551]). The results of the risk stratification study, designed to provide a new tool to aid clinical decision making for patient care, are reported on line in the journal Urology.

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 study, which analyzed tissue collected from radical prostatectomy specimens, the live tumor cell phenotypic test predicted specific post-surgical adverse pathology features – the gold standard of prostate cancer clinical diagnosis – with a high degree of sensitivity and specificity," said David Albala, MD, Chief of Urology at Crouse Hospital (Syracuse, NY) and an author on the paper. "Significantly, the test identified subgroups of prostate cancer patients within established low and intermediate Gleason and Prostate Cancer Grading Group (PGGC) tumor grades who had a higher risk based on adverse pathology features, such as positive surgical margins, lymph node involvement, and extra-prostatic extension. These initial clinical results suggest considerable potential of this phenotypic test as a risk stratification tool for prostate cancer patients with low and intermediate grade disease. The results will need to be confirmed in future studies in prostate cancer patients at the time of initial biopsy."

"Risk stratification in prostate cancer – distinguishing clinically significant cancer from indolent disease – remains a major challenge in men with low and intermediate grade disease," Dr. Albala commented. "A subset of these patients may develop aggressive disease and we currently lack sufficiently precise, personalized risk stratification tools to distinguish between indolent and potentially aggressive disease." Dr. Albala is a member of the Cellanyx Scientific Advisory Board (SAB).

Prostate cancer is the most common cancer in men in the United States, with an estimated 164,690 news cases diagnosed in 2018, according to the American Cancer Society. Although most men do not die of the disease – there will be estimated 39,430 deaths from prostate cancer in 2018. The inability of current tests to provide risk stratification of aggressive disease in low and intermediate grade patients leads to missed diagnoses, and inadequate treatment.

"Tumor heterogeneity and risk stratification are major challenges in the contemporary management of prostate cancer," said Grannum R Sant, MD, a co-author, Professor of Urology at Tufts University, Chairman of Cellanyx’s SAB and a board member. "This Cellanyx proof of concept study of the first in class, live single cell phenotypic biomarker platform is a major contribution to personalized oncology. If these findings are confirmed in a planned prostate needle biopsy trial, this phenotypic biomarker test will significantly augment Gleason, Grade Group and formalin-fixed tissue genomic analysis in risk stratification of Gleason 6 and Gleason 7 (3+4, 4+3) prostate cancer."

Study Details
The study was a multi-center, blinded, prospective trial that evaluated fresh prostate tissue samples taken from 251 men undergoing radical prostatectomy, of which 237 samples were successfully cultured and analyzed. The samples were evaluated in a central laboratory where they were tested, on a specially coated microfluidic chip and analyzed for phenotypic biomarkers in individual cells using machine vision and machine learning algorithms. The predictions of specific adverse pathology features were then compared to the actual post-surgical pathology reported findings following data un-blinding.

"The machine vision learning and intelligent algorithms developed by our team allowed objective prioritization and scoring of phenotypic biomarkers for each cell, actionable scores for predicting adverse pathological features that clinicians can use to risk-stratify patients," said Ashok Chander, PhD, an author on the paper and co-founder of Cellanyx.

The Cellanyx test accurately predicted post-radical prostatectomy adverse pathology features with an area under the curve (AUC) through receive operating characteristics analysis of greater than 0.85. The test distinguished among low and intermediate grade cancers (Gleason 3+3, 3+4 and 4+3 and PCGG 1, 2 and 3) with high precision (AUC >0.80).

The live single cell phenotypic biomarker test is being developed as a Laboratory Developed Test that can be run in any CLIA laboratory. The patient sample characteristics and acquisition were designed to fit seamlessly into the workflow of the urologist.

The paper is entitled, "Clinical proof-of-concept of a novel platform utilizing biopsy-derived live single cells, phenotypic biomarkers, and machine learning toward a precision risk stratification test for Prostate Cancer Grade Groups 1 and 2 (Gleason 3+3 and 3+4)."

Castle Biosciences to Present at the 2018 Piper Jaffray Healthcare Conference

On November 21, 2018 Castle Biosciences, Inc., a skin cancer diagnostics company providing molecular diagnostics to improve cancer management decisions, reported that Derek Maetzold, President and CEO, will present a company overview at the 30th Annual Piper Jaffray Healthcare Conference on Wednesday, November 28th, 2018 at 10:10 a.m. EST in New York City (Press release, Castle Biosciences, NOV 21, 2018, View Source [SID1234531550]).

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!


Genentech Announces FDA Grants Venclexta Accelerated Approval for People With Newly-Diagnosed Acute Myeloid Leukemia or Those Who Are Ineligible for Intensive Induction Chemotherapy

On November 21, 2018 Genentech, a member of the Roche Group (SIX: RO, ROG; OTCQX: RHHBY), reported that the U.S. Food and Drug Administration (FDA) has granted accelerated approval to Venclexta (venetoclax), in combination with a hypomethylating agent (azacitidine or decitabine), or low-dose cytarabine (LDAC), for the treatment of people with newly-diagnosed acute myeloid leukemia (AML), who are age 75 years or older, or for those ineligible for intensive induction chemotherapy due to coexisting medical conditions (Press release, Genentech, NOV 21, 2018, View Source [SID1234531549]). AML is the most common type of aggressive leukemia in adults and has the lowest survival rate for all types of leukemia.

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!

"Today’s approval marks a significant advance for people with acute myeloid leukemia, a highly aggressive and difficult-to-treat blood cancer," said Sandra Horning, M.D., chief medical officer and head of Global Product Development. "Many people with acute myeloid leukemia are unable to tolerate standard intensive chemotherapy, and the Venclexta combination regimens represent important new options for these patients."

This accelerated approval was based on results from the M14-358 study and the M14-387 study in people newly-diagnosed with AML including those who were ineligible for intensive induction chemotherapy. In M14-358, the rate of complete remission (CR) was 37 percent (n=25/67) and the rate of complete remission with partial blood count recovery (CRh) was 24 percent (n=16/67) for those who received Venclexta plus azacitidine. For those who received Venclexta plus decitabine, the rate of CR was 54 percent (n=7/13) and the rate of CRh was 8 percent (n=1/13). M14-387 showed a CR rate of 21 percent (n=13/61) and a CRh rate of 21 percent (n=13/61) for those who received Venclexta in combination with LDAC.

The most common serious side effects of these regimens (occurring in at least 5 percent of patients) were low white blood cell count with fever, pneumonia, bacteria in the blood, inflammation of tissue under the skin, device-related infection, diarrhea, fatigue, bleeding, localized infection, multiple organ dysfunction syndrome and respiratory failure.

The FDA’s Accelerated Approval Program allows conditional approval of a medicine that fills an unmet medical need for a serious condition. This approval of Venclexta is based on surrogate endpoints that are reasonably likely to predict clinical benefit, including CR and CRh. Continued approval for this indication may be contingent upon verification and description of clinical benefit observed in confirmatory trials.

The supplemental New Drug Application (sNDA) was granted Priority Review, a designation given to medicines that the FDA has determined to have the potential to provide significant improvements in the treatment, prevention or diagnosis of a disease. In addition, the FDA previously granted two Breakthrough Therapy Designations for Venclexta in people with previously untreated AML ineligible for intensive chemotherapy, either in combination with a hypomethylating agent or LDAC, based on results from these two studies. With this approval, Venclexta is available in the U.S. for two forms of blood cancer.

Venclexta is being developed by AbbVie and Genentech, a member of the Roche Group. It is jointly commercialized by the companies in the U.S. and commercialized by AbbVie outside of the U.S.

About the M14-358 study

The M14-358 study (NCT02203773) is an open-label, non-randomized, Phase Ib dose escalation and expansion study evaluating the safety and efficacy of Venclexta in combination with hypomethylating agents, azacitidine or decitabine, in newly-diagnosed people with AML who were 60 years or older, or ineligible to receive intensive induction chemotherapy due to coexisting medical conditions. Study endpoints included complete remission rates, overall survival and safety.

In M14-358, the rate of CR was 37 percent and the rate of CRh was 24 percent for those who received Venclexta plus azacitidine. The median follow-up for this group was 7.9 months (0.4-36 months). At the time of analysis, for patients who achieved a CR, the median observed time in remission was 5.5 months (0.4-30 months).
For those who received Venclexta plus decitabine, the rate of CR was 54 percent and the rate of CRh was 8 percent. The median follow-up for this group was 11 months (0.7-21 months). At the time of analysis, for patients who achieved a CR, the median observed time in remission was 4.7 months (1.0-18 months).
The observed time in remission for these regimens was defined as the time from the start of CR to the time of the data cut-off date or relapse from CR.
The most common adverse reactions with Venclexta plus azacitidine were nausea, diarrhea, constipation, low white blood cell count with or without fever, low platelet count, bleeding, swelling in the arms, legs, hands and feet, vomiting, fatigue, rash and low red blood cell count.
The most common adverse reactions with Venclexta plus decitabine were low white blood cell count with or without fever, constipation, fatigue, low platelet count, stomach (abdominal) pain, dizziness, bleeding, nausea, pneumonia, infection in the blood, cough, diarrhea, low blood pressure, pain in muscles or back, sore throat, swelling in the arms, legs, hands and feet, fever and rash.
About the M14-387 study

The M14-387 study (NCT02287233) is an open-label, single-arm, Phase I/II dose escalation and expansion study evaluating the safety and efficacy of Venclexta in combination with LDAC in newly-diagnosed people with AML who were 60 years or older, or ineligible to receive intensive induction chemotherapy due to coexisting medical conditions. Study endpoints included complete remission rates, overall survival and safety.

The study showed the rate of CR and CRh was 21 percent for those who received Venclexta plus LDAC. The median follow-up for this group was 6.5 months (0.3-34 months). At the time of analysis, for patients who achieved a CR, the median observed time in remission was 6.0 months (0.03-25 months). The observed time in remission for this regimen was defined as the time from the start of CR to the time of the data cut-off date or relapse from CR.
The most common adverse reactions with Venclexta in combination with LDAC were nausea, low platelet count, bleeding, low white blood cell count with or without fever, diarrhea, fatigue, constipation and difficulty breathing.
About AML

Acute myeloid leukemia (AML) is the most common type of aggressive leukemia in adults, which has the lowest survival rate for all types of leukemia. In 2018, it is estimated there will be nearly 20,000 new cases of AML diagnosed in the United States. Many AML patients older than age 60 are unable to tolerate intensive induction chemotherapy treatment.

About Genentech Access Solutions

Access Solutions is part of Genentech’s commitment to helping people access the Genentech medicines they are prescribed, regardless of their ability to pay. The team of in-house specialists at Access Solutions is dedicated to helping people navigate the access and reimbursement process, and to providing assistance to eligible patients in the United States who are uninsured or cannot afford the out-of-pocket costs for their medicine. To date, the team has helped more than 1.5 million patients access the medicines they need. Please contact Access Solutions (866) 4ACCESS/(866) 422-2377 or visit View Source for more information.

About Venclexta

Venclexta is a first-in-class targeted medicine designed to selectively bind and inhibit the B-cell lymphoma-2 (BCL-2) protein. In some blood cancers and other tumors, BCL-2 builds up and prevents cancer cells from dying or self-destructing, a process called apoptosis. Venclexta blocks the BCL-2 protein and works to restore the process of apoptosis.

Venclexta is being developed by AbbVie and Genentech, a member of the Roche Group. It is jointly commercialized by the companies in the U.S. and commercialized by AbbVie outside of the U.S. Together, the companies are committed to research with Venclexta, which is currently being studied in clinical trials across several types of blood and other cancers.

In the U.S., Venclexta has been granted four Breakthrough Therapy Designations by the FDA: in combination with Rituxan for people with relapsed or refractory chronic lymphocytic leukemia (CLL); as a monotherapy for people with relapsed or refractory CLL with 17p deletion; in combination with hypomethylating agents (azacitidine or decitabine) for people with untreated acute myeloid leukemia (AML) ineligible for intensive chemotherapy; and in combination with low-dose cytarabine for people with untreated AML ineligible for intensive chemotherapy.

Venclexta Indications

Venclexta is a prescription medicine used:

To treat adults with chronic lymphocytic leukemia (CLL) or small lymphocytic lymphoma (SLL), with or without 17p deletion, who have received at least 1 prior treatment.
In combination with azacitidine, or decitabine, or low-dose cytarabine to treat adults with newly-diagnosed acute myeloid leukemia (AML) who:
‒ Are 75 years of age or older, or
‒ Have other medical conditions that prevent the use of standard chemotherapy.
It is not known if Venclexta is safe and effective in children.

Important Safety Information

Venclexta can cause serious side effects, including:

Tumor lysis syndrome (TLS). TLS is caused by the fast breakdown of cancer cells. TLS can cause kidney failure, the need for dialysis treatment, and may lead to death. The patient’s doctor will do tests to check their risk of getting TLS before they start taking Venclexta. The patient will receive other medicines before starting and during treatment with Venclexta to help reduce the risk of TLS. The patient may also need to receive intravenous (IV) fluids through their vein.

The patient’s doctor will do blood tests to check for TLS when the patient first starts treatment and during treatment with Venclexta. It is important for patients to keep appointments for blood tests. Patients should tell their doctor right away if they have any symptoms of TLS during treatment with Venclexta, including fever, chills, nausea, vomiting, confusion, shortness of breath, seizures, irregular heartbeat, dark or cloudy urine, unusual tiredness, or muscle or joint pain.

Patients should drink plenty of water during treatment with Venclexta to help reduce the risk of getting TLS.

Patients should drink 6 to 8 glasses (about 56 ounces total) of water each day, starting 2 days before the first dose, on the day of the first dose of Venclexta, and each time a dose is increased.

The patient’s doctor may delay, decrease the dose, or stop treatment with Venclexta if the patient has side effects.

Certain medicines must not be taken when the patient first starts taking Venclexta and while the dose is being slowly increased because of the risk of increased tumor lysis syndrome.

Patients must tell their doctor about all the medicines they take, including prescription and over-the-counter medicines, vitamins, and herbal supplements. Venclexta and other medicines may affect each other, causing serious side effects.
Patients must not start new medicines during treatment with Venclexta without first talking with their doctor.
Before taking Venclexta, patients must tell their doctor about all of their medical conditions, including if they:

Have kidney problems.
Have problems with body salts or electrolytes, such as potassium, phosphorus, or calcium.
Have a history of high uric acid levels in the blood or gout.
Are scheduled to receive a vaccine. The patient should not receive a "live vaccine" before, during, or after treatment with Venclexta, until the patient’s doctor tells them it is okay. If the patient is not sure about the type of immunization or vaccine, the patient should ask their doctor. These vaccines may not be safe or may not work as well during treatment with Venclexta.
Are pregnant or plan to become pregnant. Venclexta may harm an unborn baby. If the patient is able to become pregnant, the patient’s doctor should do a pregnancy test before the patient starts treatment with Venclexta, and the patient should use effective birth control during treatment and for at least 30 days after the last dose of Venclexta. If the patient becomes pregnant or thinks they are pregnant, the patient should tell their doctor right away.
Are breastfeeding or plan to breastfeed. It is not known if Venclexta passes into the patient’s breast milk. Patients should not breastfeed during treatment with Venclexta.
What to avoid while taking Venclexta:

Patients should not drink grapefruit juice, eat grapefruit, Seville oranges (often used in marmalades), or starfruit while they are taking Venclexta. These products may increase the amount of Venclexta in the patient’s blood.

Venclexta can cause serious side effects, including:

Low white blood cell counts (neutropenia). Low white blood cell counts are common with Venclexta, but can also be severe. The patient’s doctor will do blood tests to check their blood counts during treatment with Venclexta. Patients should tell their doctor right away if they have a fever or any signs of an infection during treatment with Venclexta.
The most common side effects of Venclexta when used in combination with rituximab in people with CLL include low white blood cell counts; diarrhea; upper respiratory tract infection; cough; tiredness; and nausea.

The most common side effects of Venclexta when used alone in people with CLL/SLL include low white blood cell counts; diarrhea; nausea; upper respiratory tract infection; low red blood cell counts; tiredness; low platelet counts; muscle and joint pain; swelling of arms, legs, hands, and feet; and cough.

The most common side effects of Venclexta in combination with azacitidine, or decitabine, or low-dose cytarabine in people with AML include low white blood cell counts; nausea; diarrhea; low platelet counts; constipation; fever with low white blood cell counts; low red blood cell counts; infection in blood; rash; dizziness; low blood pressure; fever; swelling of arms, legs, hands, and feet; vomiting; tiredness; shortness of breath; bleeding; infection in lung; stomach (abdominal) pain; pain in muscles or back; cough; and sore throat.

Venclexta may cause fertility problems in males. This may affect the ability to father a child. Patients should talk to their doctor if they have concerns about fertility.

These are not all the possible side effects of Venclexta. Patients should tell their doctor about any side effect that bothers them or that does not go away.

U.S. FDA Approves DAURISMO™ (glasdegib) for Adult Patients with Newly-Diagnosed Acute Myeloid Leukemia (AML) for Whom Intensive Chemotherapy is Not an Option

On November 21, 2018 Pfizer Inc. (NYSE:PFE) reported that the U.S. Food and Drug Administration (FDA) approved DAURISMO (glasdegib), a once-daily oral medicine, for the treatment of newly-diagnosed acute myeloid leukemia (AML) in adult patients who are 75 years or older or who have comorbidities that preclude use of intensive induction chemotherapy (Press release, Pfizer, NOV 21, 2018, View Source [SID1234531548]). DAURISMO is taken in combination with low-dose cytarabine (LDAC), a type of chemotherapy. DAURISMO has not been studied in patients with severe renal impairment or moderate-to-severe hepatic impairment.1

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!

AML is a rapidly progressing bone marrow cancer with poor survival rates compared to other leukemias.2 The standard of care for people with AML is intensive chemotherapy; however, for many elderly patients with AML, as well as those who have certain health conditions prior to receiving their diagnosis, intensive treatment is not an option.3 Historically, a majority of these individuals do not receive treatment and face a poor prognosis.4

"As our second medicine approved in the last 14 months for patients with acute myeloid leukemia, DAURISMO reinforces our commitment to delivering new medicines to patients living with some of the most difficult-to-treat cancers, especially those for which there are limited treatment options available," said Andy Schmeltz, Global President, Pfizer Oncology. "We are proud to now offer these patients for whom intensive chemotherapy is not an option a new oral medicine, taken in combination with low-dose chemotherapy, that may improve their chances of survival."

DAURISMO is the first and only FDA-approved Hedgehog pathway inhibitor for AML. The Hedgehog signaling pathway plays an essential role in embryogenesis, the process by which human embryos are developed. In adults, however, abnormal activation of this pathway is thought to contribute to the development and persistence of cancer stem cells. Preclinical studies have shown that disruption of this pathway can impair the development and survival of these cancer stem cells.5,6

"The randomized Phase 2 study, which formed the basis for today’s approval, included patients with cardiac disease or mild to moderate kidney disease, who are often excluded from clinical trials," said Jorge Cortes, M.D., deputy chair and professor of medicine in the Department of Leukemia, University of Texas, MD Anderson Cancer Center. "In the trial, DAURISMO plus low-dose chemotherapy reduced the risk of death during the study period by 54 percent compared to chemotherapy alone. This provides a much-needed treatment for those patients for whom intensive chemotherapy is not an option."

In the pivotal, randomized, international Phase 2 BRIGHT 1003 trial, 115 patients with newly diagnosed AML were randomized 2:1 to receive DAURISMO plus LDAC or LDAC alone. Of the 77 patients treated with DAURISMO plus LDAC, more than half (51%, 39 patients) had secondary AML, or AML that develops as a result of prior blood/bone marrow conditions or previous anticancer therapy. Eleven of the 39 patients with secondary AML received prior treatment with a hypomethylating agent; historically, the prognosis is poor for these patients and treatment options have been limited to clinical trials or palliative care. Median overall survival was 8.3 months (95% CI: 4.4,12.2) for patients treated with DAURISMO plus LDAC compared with 4.3 months (95% CI: 1.9, 5.7) for patients treated with LDAC alone. This difference represented a 54 percent reduction in the risk of death for patients treated with DAURISMO plus LDAC (HR: 0.46, 95% CI: 0.30, 0.71, one-sided p-value 0.0002).1

The U.S. labeling for DAURISMO includes a boxed warning for embryo-fetal toxicity. The most frequently (≥20% of patients) reported adverse events (AEs) in patients treated with DAURISMO plus LDAC compared to LDAC alone in first 90 days of therapy were anemia (43% vs 42%), fatigue (36% vs 32%), hemorrhage (36% vs 42%), febrile neutropenia (31% vs 22%), musculoskeletal pain (30% vs 17%), nausea (29% vs 12%), edema (30% vs 20%), thrombocytopenia (30% vs 27%), dyspnea (23% vs 24%), decreased appetite (21% vs 7%), dysgeusia (21% vs 2%), mucositis (21% vs 12%), constipation (20% vs 12%) and rash (20% vs 7%).1 Serious adverse reactions were reported in 79% of patients treated in the DAURISMO plus LDAC arm. The most common (≥5%) serious adverse reactions in patients receiving DAURISMO plus LDAC were febrile neutropenia (29%), pneumonia (23%), hemorrhage (12%), anemia (7%) and sepsis (7%).1

"DAURISMO, a Hedgehog pathway inhibitor, was discovered in Pfizer laboratories and exemplifies our continued commitment to developing medicines that have the potential to advance cancer therapeutics," said Mace Rothenberg, M.D., Chief Development Officer, Oncology, Pfizer Global Product Development. "We are delighted by today’s approval of DAURISMO by the FDA, and are working to gain greater understanding of its role in treating patients with acute myeloid leukemia. The ongoing Phase 3 BRIGHT trials are evaluating DAURISMO in combination with other agents commonly used to treat patients with acute myeloid leukemia, in an effort to understand the full potential of this medicine against this aggressive leukemia."

Pfizer is committed to ensuring that patients who are prescribed DAURISMO have access to this innovative therapy. Patients in the U.S. have access to Pfizer Oncology Together, which offers personalized support and financial assistance resources to help patients access their prescribed Pfizer Oncology medications.

The full Prescribing Information, including BOXED WARNING, for DAURISMO can be found here.

IMPORTANT DAURISMO SAFETY INFORMATION FROM THE U.S. PRESCRIBING INFORMATION

WARNING: EMBRYO-FETAL TOXICITY: DAURISMO can cause embryo-fetal death or severe birth defects when administered to a pregnant woman. DAURISMO is embryotoxic, fetotoxic, and teratogenic in animals. Conduct pregnancy testing in females of reproductive potential prior to initiation of DAURISMO treatment. Advise females of reproductive potential to use effective contraception during treatment with DAURISMO and for at least 30 days after the last dose. Advise males of the potential risk of DAURISMO exposure through semen and to use condoms with a pregnant partner or a female partner of reproductive potential during treatment with DAURISMO and for at least 30 days after the last dose to avoid potential drug exposure.

Blood Donation: Advise patients not to donate blood or blood products while taking DAURISMO and for at least 30 days after the last dose, because their blood or blood products might be given to a female of reproductive potential.

QTc Interval Prolongation: Patients treated with DAURISMO can develop QTc prolongation and ventricular arrhythmias, including ventricular fibrillation and ventricular tachycardia. Monitor electrocardiograms (ECGs) and electrolytes. Concomitant use of DAURISMO with drugs known to prolong the QTc interval and CYP3A4 inhibitors may increase the risk of QTc interval prolongation. In patients with congenital long QT syndrome, congestive heart failure, electrolyte abnormalities, or those who are taking medications known to prolong the QTc interval, more frequent ECG monitoring is recommended. Interrupt DAURISMO if QTc interval is >500 ms and discontinue permanently for patients who develop QTc interval prolongation with signs or symptoms of life-threatening arrhythmia.

Adverse Reactions: Most common adverse reactions (incidence ≥20%) are anemia, fatigue, hemorrhage, febrile neutropenia, musculoskeletal pain, nausea, edema, thrombocytopenia, dyspnea, decreased appetite, dysgeusia, mucositis, constipation, and rash.

Drug Interactions: Co-administration with strong CYP3A4 inhibitors increased DAURISMO plasma concentrations, which may increase the risk of adverse reactions including QTc interval prolongation. Consider alternative therapies that are not strong CYP3A4 inhibitors during treatment with DAURISMO and monitor patients for increased risk of adverse reactions including QTc interval prolongation. Strong CYP3A4 inducers should be avoided due to decreased DAURISMO plasma concentrations, which may reduce efficacy.

Lactation: Because of the potential for serious adverse reactions from DAURISMO in a breastfed child, advise women who are taking DAURISMO not to breastfeed or provide breast milk to infants or children during treatment and for at least 30 days after the last dose.

About DAURISMO (glasdegib)

DAURISMO is a once-daily oral Hedgehog pathway inhibitor, taken in combination with LDAC, for the treatment of newly diagnosed AML in adult patients who are 75 years or older or who have comorbidities that preclude use of intensive induction chemotherapy.1 DAURISMO has not been studied in patients with severe renal impairment or moderate-to-severe hepatic impairment. As an oral therapy, which is taken with subcutaneous LDAC, DAURISMO offers the flexibility for patients to receive this treatment regimen at home or in the outpatient setting.

DAURISMO was discovered in Pfizer’s U.S. laboratories and we utilize state-of-the-art continuous manufacturing to produce this treatment.

DAURISMO is not approved for any indication in any market outside the U.S.

About the BRIGHT Clinical Trials

BRIGHT AML 1019 (NCT03416179) consists of two randomized, placebo-controlled Phase 3 trials evaluating the addition of DAURISMO to intensive or non-intensive chemotherapy in patients with newly diagnosed AML. In the first study, patients with AML will be randomized to receive DAURISMO plus cytarabine and daunorubicin, an intensive chemotherapy regimen, or placebo plus cytarabine and daunorubicin. In the second study, patients with AML for whom intensive chemotherapy is not an option will be randomized to receive DAURISMO plus azacitidine, a hypomethylating agent, or placebo plus azacitidine.

A separate Phase 1b BRIGHT 1012 study (NCT02367456) has also been expanded to evaluate DAURISMO in combination with azacitidine in patients with previously untreated high-risk myelodysplastic syndromes (MDS) or AML. These trials are currently enrolling patients.

About Pfizer Hematology

Pfizer’s commitment to hematologic malignancies began in 2012 with the approval of our treatment for chronic myeloid leukemia (CML). Since then, we’ve continued to expand our hematology portfolio to meet the needs of patients with acute lymphoblastic leukemia (ALL) and AML. We now have four products approved for leukemia in different countries around the world, including three in the past two years. Together with the community, Pfizer aims to overcome the challenges of hematologic cancers and translate breakthrough science into meaningful treatment advances for patients.