Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Philip Friedlander is active.

Publication


Featured researches published by Philip Friedlander.


The New England Journal of Medicine | 2012

Efficacy and Safety of Vismodegib in Advanced Basal-Cell Carcinoma

Aleksandar Sekulic; Michael R. Migden; Anthony E. Oro; Luc Dirix; Karl D. Lewis; John D. Hainsworth; James A. Solomon; Simon Yoo; Sarah T. Arron; Philip Friedlander; Ellen S. Marmur; Charles M. Rudin; Anne Lynn S. Chang; Jennifer A. Low; Howard Mackey; Robert L. Yauch; Richard A. Graham; Josina C. Reddy; Axel Hauschild

BACKGROUND Alterations in hedgehog signaling are implicated in the pathogenesis of basal-cell carcinoma. Although most basal-cell carcinomas are treated surgically, no effective therapy exists for locally advanced or metastatic basal-cell carcinoma. A phase 1 study of vismodegib (GDC-0449), a first-in-class, small-molecule inhibitor of the hedgehog pathway, showed a 58% response rate among patients with advanced basal-cell carcinoma. METHODS In this multicenter, international, two-cohort, nonrandomized study, we enrolled patients with metastatic basal-cell carcinoma and those with locally advanced basal-cell carcinoma who had inoperable disease or for whom surgery was inappropriate (because of multiple recurrences and a low likelihood of surgical cure, or substantial anticipated disfigurement). All patients received 150 mg of oral vismodegib daily. The primary end point was the independently assessed objective response rate; the primary hypotheses were that the response rate would be greater than 20% for patients with locally advanced basal-cell carcinoma and greater than 10% for those with metastatic basal-cell carcinoma. RESULTS In 33 patients with metastatic basal-cell carcinoma, the independently assessed response rate was 30% (95% confidence interval [CI], 16 to 48; P=0.001). In 63 patients with locally advanced basal-cell carcinoma, the independently assessed response rate was 43% (95% CI, 31 to 56; P<0.001), with complete responses in 13 patients (21%). The median duration of response was 7.6 months in both cohorts. Adverse events occurring in more than 30% of patients were muscle spasms, alopecia, dysgeusia (taste disturbance), weight loss, and fatigue. Serious adverse events were reported in 25% of patients; seven deaths due to adverse events were noted. CONCLUSIONS Vismodegib is associated with tumor responses in patients with locally advanced or metastatic basal-cell carcinoma. (Funded by Genentech; Erivance BCC ClinicalTrials.gov number, NCT00833417.).


Journal of Clinical Oncology | 2008

Major Response to Imatinib Mesylate in KIT-Mutated Melanoma

F. Stephen Hodi; Philip Friedlander; Christopher L. Corless; Michael C. Heinrich; Suzanne Mac Rae; Andrea Kruse; Jyothi P. Jagannathan; Annick D. Van den Abbeele; Elsa F. Velazquez; George D. Demetri; David E. Fisher

addition, this is the first reported case of a synchronous malignant melanoma and a small-cell lung cancer metastasizing to the same anatomic location. This case illustrates the necessity of proper pathologic evaluation in a brain lesion assumed to be a metastasis. In this case, had the patient’s performance status improved after her motor vehicle accident, she would have been offered systemic chemotherapy for her extensive-stage smallcell lung cancer, which can markedly improve survival, even in advanced disease. If this brain lesion had proven to be solely melanoma, it is unlikely that any systemic treatment would have been offered.


The New England Journal of Medicine | 2016

PD-1 Blockade with Pembrolizumab in Advanced Merkel-Cell Carcinoma

Paul Nghiem; Shailender Bhatia; Evan J. Lipson; Ragini R. Kudchadkar; Natalie J. Miller; Lakshmanan Annamalai; Sneha Berry; Elliot Chartash; Adil Daud; Steven P. Fling; Philip Friedlander; Harriet M. Kluger; Holbrook Kohrt; Lisa Lundgren; Kim Margolin; Alan Mitchell; Thomas Olencki; Drew M. Pardoll; Sunil Reddy; Erica Shantha; William H. Sharfman; Elad Sharon; Lynn R. Shemanski; Michi M. Shinohara; Joel C. Sunshine; Janis M. Taube; John A. Thompson; Steven M. Townson; Jennifer H. Yearley; Suzanne L. Topalian

BACKGROUND Merkel-cell carcinoma is an aggressive skin cancer that is linked to exposure to ultraviolet light and the Merkel-cell polyomavirus (MCPyV). Advanced Merkel-cell carcinoma often responds to chemotherapy, but responses are transient. Blocking the programmed death 1 (PD-1) immune inhibitory pathway is of interest, because these tumors often express PD-L1, and MCPyV-specific T cells express PD-1. METHODS In this multicenter, phase 2, noncontrolled study, we assigned adults with advanced Merkel-cell carcinoma who had received no previous systemic therapy to receive pembrolizumab (anti-PD-1) at a dose of 2 mg per kilogram of body weight every 3 weeks. The primary end point was the objective response rate according to Response Evaluation Criteria in Solid Tumors, version 1.1. Efficacy was correlated with tumor viral status, as assessed by serologic and immunohistochemical testing. RESULTS A total of 26 patients received at least one dose of pembrolizumab. The objective response rate among the 25 patients with at least one evaluation during treatment was 56% (95% confidence interval [CI], 35 to 76); 4 patients had a complete response, and 10 had a partial response. With a median follow-up of 33 weeks (range, 7 to 53), relapses occurred in 2 of the 14 patients who had had a response (14%). The response duration ranged from at least 2.2 months to at least 9.7 months. The rate of progression-free survival at 6 months was 67% (95% CI, 49 to 86). A total of 17 of the 26 patients (65%) had virus-positive tumors. The response rate was 62% among patients with MCPyV-positive tumors (10 of 16 patients) and 44% among those with virus-negative tumors (4 of 9 patients). Drug-related grade 3 or 4 adverse events occurred in 15% of the patients. CONCLUSIONS In this study, first-line therapy with pembrolizumab in patients with advanced Merkel-cell carcinoma was associated with an objective response rate of 56%. Responses were observed in patients with virus-positive tumors and those with virus-negative tumors. (Funded by the National Cancer Institute and Merck; ClinicalTrials.gov number, NCT02267603.).


Journal of Clinical Oncology | 2013

Imatinib for Melanomas Harboring Mutationally Activated or Amplified KIT Arising on Mucosal, Acral, and Chronically Sun-Damaged Skin

F. Stephen Hodi; Christopher L. Corless; Anita Giobbie-Hurder; Jonathan A. Fletcher; Meijun Zhu; Adrián Mariño-Enríquez; Philip Friedlander; Rene Gonzalez; Jeffrey S. Weber; Thomas F. Gajewski; Steven O'Day; Kevin B. Kim; Donald P. Lawrence; Keith T. Flaherty; Jason J. Luke; Frances A. Collichio; Marc S. Ernstoff; Michael C. Heinrich; Carol Beadling; Katherine Zukotynski; Jeffrey T. Yap; Annick D. Van den Abbeele; George D. Demetri; David E. Fisher

PURPOSE Amplifications and mutations in the KIT proto-oncogene in subsets of melanomas provide therapeutic opportunities. PATIENTS AND METHODS We conducted a multicenter phase II trial of imatinib in metastatic mucosal, acral, or chronically sun-damaged (CSD) melanoma with KIT amplifications and/or mutations. Patients received imatinib 400 mg once per day or 400 mg twice per day if there was no initial response. Dose reductions were permitted for treatment-related toxicities. Additional oncogene mutation screening was performed by mass spectroscopy. RESULTS Twenty-five patients were enrolled (24 evaluable). Eight patients (33%) had tumors with KIT mutations, 11 (46%) with KIT amplifications, and five (21%) with both. Median follow-up was 10.6 months (range, 3.7 to 27.1 months). Best overall response rate (BORR) was 29% (21% excluding nonconfirmed responses) with a two-stage 95% CI of 13% to 51%. BORR was significantly greater than the hypothesized null of 5% and statistically significantly different by mutation status (7 of 13 or 54% KIT mutated v 0% KIT amplified only). There were no statistical differences in rates of progression or survival by mutation status or by melanoma site. The overall disease control rate was 50% but varied significantly by KIT mutation status (77% mutated v 18% amplified). Four patients harbored pretreatment NRAS mutations, and one patient acquired increased KIT amplification after treatment. CONCLUSION Melanomas that arise on mucosal, acral, or CSD skin should be assessed for KIT mutations. Imatinib can be effective when tumors harbor KIT mutations, but not if KIT is amplified only. NRAS mutations and KIT copy number gain may be mechanisms of therapeutic resistance to imatinib.


Cancer immunology research | 2014

Bevacizumab plus Ipilimumab in Patients with Metastatic Melanoma

F. Stephen Hodi; Donald P. Lawrence; Cecilia Lezcano; Xinqi Wu; Jun Zhou; Tetsuro Sasada; Wanyong Zeng; Anita Giobbie-Hurder; Michael B. Atkins; Nageatte Ibrahim; Philip Friedlander; Keith T. Flaherty; George F. Murphy; Scott J. Rodig; Elsa F. Velazquez; Martin C. Mihm; Sara Russell; Pamela J. DiPiro; Jeffrey T. Yap; Nikhil Ramaiya; Annick D. Van den Abbeele; Maria Gargano; David F. McDermott

Hodi and colleagues report the safety and efficacy of targeting angiogenesis and CTLA-4 in a phase I trial of 46 patients with metastatic melanoma, which revealed the influence of VEGF-A blockade on inflammation, lymphocyte trafficking, and immune regulation, and their synergistic therapeutic effects. Ipilimumab improves survival in advanced melanoma and can induce immune-mediated tumor vasculopathy. Besides promoting angiogenesis, vascular endothelial growth factor (VEGF) suppresses dendritic cell maturation and modulates lymphocyte endothelial trafficking. This study investigated the combination of CTLA4 blockade with ipilimumab and VEGF inhibition with bevacizumab. Patients with metastatic melanoma were treated in four dosing cohorts of ipilimumab (3 or 10 mg/kg) with four doses at 3-week intervals and then every 12 weeks, and bevacizumab (7.5 or 15 mg/kg) every 3 weeks. Forty-six patients were treated. Inflammatory events included giant cell arteritis (n = 1), hepatitis (n = 2), and uveitis (n = 2). On-treatment tumor biopsies revealed activated vessel endothelium with extensive CD8+ and macrophage cell infiltration. Peripheral blood analyses demonstrated increases in CCR7+/−/CD45RO+ cells and anti-galectin antibodies. Best overall response included 8 partial responses, 22 instances of stable disease, and a disease-control rate of 67.4%. Median survival was 25.1 months. Bevacizumab influences changes in tumor vasculature and immune responses with ipilimumab administration. The combination of bevacizumab and ipilimumab can be safely administered and reveals VEGF-A blockade influences on inflammation, lymphocyte trafficking, and immune regulation. These findings provide a basis for further investigating the dual roles of angiogenic factors in blood vessel formation and immune regulation, as well as future combinations of antiangiogenesis agents and immune checkpoint blockade. Cancer Immunol Res; 2(7); 632–42. ©2014 AACR.


Science Translational Medicine | 2011

Establishment of antitumor memory in humans using in vitro-educated CD8+ T cells.

Marcus O. Butler; Philip Friedlander; Matthew I. Milstein; Mary M. Mooney; Genita Metzler; Andrew P. Murray; Makito Tanaka; Alla Berezovskaya; Osamu Imataki; Linda Drury; Lisa Brennan; Marisa Flavin; Donna Neuberg; Kristen E. Stevenson; Donald P. Lawrence; F. Stephen Hodi; Elsa F. Velazquez; Michael T. Jaklitsch; Sara Russell; Martin C. Mihm; Lee M. Nadler; Naoto Hirano

Antitumor CD8+ T cells educated in vitro can persist as memory T cells and induce antitumor responses in humans without prior conditioning or cytokine treatment. Memory that Keeps Going and Going Senior scientists consoling their trainees about failed experiments tout the value of persistence. Yet, persistence is not only important for the scientist tirelessly pipetting at two in the morning, it is key for immunological memory as well. Melanoma is a malignant tumor of melanocytes, the pigment cells found in the skin. Advanced-stage melanoma has a poor prognosis, with patients on average surviving less than a year. A new promising therapy for advanced-stage melanoma patients harnesses the immune system to attack the tumor cells. In adoptive T cell therapy, cytotoxic cells specific for the tumor are transferred into patients, where they then traffic to and destroy the tumor cells. However, one limitation of this therapy is keeping the tumor-specific T cells alive in the patient, which has been largely unsuccessful in the absence of extensive treatment of the patient. Butler et al. now report a means to expand these T cells that allows them to persist in the absence of extra patient manipulation. The authors use artificial antigen-presenting cells to turn antitumor T cells into memory T cells, which survive longer and respond more quickly than normal effector T cells. These artificial antigen-presenting cells express molecules that “costimulate” the T cells, resulting in T cells that both look and act like memory T cells in the culture dish. These educated tumor-specific cells were then introduced into patients with advanced-stage melanoma. These cells functioned as memory cells in the patients as well: persisting for long periods of time, homing to the tumor site, and demonstrating tumor-specific activation and function, all in the absence of further patient manipulation. Although this is an early clinical trial with a small number of patients, there is some indication that this treatment may have clinical benefit for patients with this devastating disease. Persistence, both of the T cells and the scientists running the study, has finally paid off. Although advanced-stage melanoma patients have a median survival of less than a year, adoptive T cell therapy can induce durable clinical responses in some patients. Successful adoptive T cell therapy to treat cancer requires engraftment of antitumor T lymphocytes that not only retain specificity and function in vivo but also display an intrinsic capacity to survive. To date, adoptively transferred antitumor CD8+ T lymphocytes (CTLs) have had limited life spans unless the host has been manipulated. To generate CTLs that have an intrinsic capacity to persist in vivo, we developed a human artificial antigen-presenting cell system that can educate antitumor CTLs to acquire both a central memory and an effector memory phenotype as well as the capacity to survive in culture for prolonged periods of time. We examined whether antitumor CTLs generated using this system could function and persist in patients. We showed that MART1-specific CTLs, educated and expanded using our artificial antigen-presenting cell system, could survive for prolonged periods in advanced-stage melanoma patients without previous conditioning or cytokine treatment. Moreover, these CTLs trafficked to the tumor, mediated biological and clinical responses, and established antitumor immunologic memory. Therefore, this approach may broaden the availability of adoptive cell therapy to patients both alone and in combination with other therapeutic modalities.


Journal of The American Academy of Dermatology | 2015

Pivotal ERIVANCE basal cell carcinoma (BCC) study: 12-month update of efficacy and safety of vismodegib in advanced BCC

Aleksandar Sekulic; Michael R. Migden; Karl D. Lewis; John D. Hainsworth; James A. Solomon; Simon Yoo; Sarah T. Arron; Philip Friedlander; Ellen S. Marmur; Charles M. Rudin; Anne Lynn S. Chang; Luc Dirix; Jeannie Hou; Huibin Yue; Axel Hauschild

BACKGROUND Primary analysis from the pivotal ERIVANCE BCC study resulted in approval of vismodegib, a Hedgehog pathway inhibitor indicated for treatment of adults with metastatic or locally advanced basal cell carcinoma (BCC) that has recurred after surgery or for patients who are not candidates for surgery or radiation. OBJECTIVE An efficacy and safety analysis was conducted 12 months after primary analysis. METHODS This was a multinational, multicenter, nonrandomized, 2-cohort study in patients with measurable and histologically confirmed locally advanced or metastatic BCC taking oral vismodegib (150 mg/d). Primary outcome measure was objective response rate (complete and partial responses) assessed by independent review facility. RESULTS After 12 months of additional follow-up, median duration of exposure to vismodegib was 12.9 months. Objective response rate increased from 30.3% to 33.3% in patients with metastatic disease, and from 42.9% to 47.6% in patients with the locally advanced form. Median duration of response in patients with locally advanced BCC increased from 7.6 to 9.5 months. No new safety signals emerged with extended treatment duration. LIMITATIONS Limitations include low prevalence of advanced BCC and challenges of designing a study with heterogenous manifestations. CONCLUSION The 12-month update of the study confirms the efficacy and safety of vismodegib in management of advanced BCC.


Journal of Clinical Oncology | 2013

Final Results of Phase III SYMMETRY Study: Randomized, Double-Blind Trial of Elesclomol Plus Paclitaxel Versus Paclitaxel Alone As Treatment for Chemotherapy-Naive Patients With Advanced Melanoma

Steven O'Day; Alexander Eggermont; Vanna Chiarion-Sileni; Richard F. Kefford; Jean Jacques Grob; Laurent Mortier; Caroline Robert; Jacob Schachter; Alessandro Testori; Jacek Mackiewicz; Philip Friedlander; Claus Garbe; Selma Ugurel; Frances A. Collichio; Wei Guo; Joelle Lufkin; Safi R. Bahcall; Vojo Vukovic; Axel Hauschild

PURPOSE Elesclomol, an investigational first-in-class compound, induces oxidative stress, triggers mitochondrial-induced apoptosis in cancer cells, and shows synergy with taxanes in tumor models. Following completion of a phase II trial of elesclomol in combination with paclitaxel that met its primary end point of progression-free survival (PFS), this randomized, double-blind, controlled phase III study was conducted to confirm the efficacy and tolerability of elesclomol in combination with paclitaxel versus paclitaxel alone in patients with advanced melanoma. PATIENTS AND METHODS Patients with stage IV chemotherapy-naive melanoma (n = 651) were randomly assigned 1:1 to paclitaxel 80 mg/m(2) either alone or in combination with elesclomol 213 mg/m(2) administered weekly for 3 weeks of a 4-week cycle. Patients were stratified by prior systemic treatment, M1 subclass, and baseline lactate dehydrogenase (LDH) levels. The primary end point was PFS. RESULTS The study did not achieve its PFS end point (hazard ratio, 0.89; P = .23). The study was stopped when an early overall survival data analysis indicated an imbalance in total deaths favoring paclitaxel, predominantly in patients with high LDH levels. A prospectively defined subgroup analysis revealed a statistically significant improvement in median PFS for the combination in patients with normal baseline LDH. CONCLUSION The addition of elesclomol to paclitaxel did not significantly improve PFS in unselected patients with advanced melanoma. The association between baseline LDH and clinical outcomes suggests that LDH may be a predictive factor for treatment with this combination, consistent with recent findings on the association between elesclomol anticancer activity and cellular metabolic state.


Melanoma Research | 2013

Sweet's syndrome in a patient with metastatic melanoma after ipilimumab therapy.

Sofya Pintova; Harleen Sidhu; Philip Friedlander; Randall F. Holcombe

Sweets syndrome, a neutrophilic dermatosis, is a known paraneoplastic complication occurring with various malignancies. It has been infrequently reported in association with melanoma. Ipilimumab is an antibody against an inhibitory cytotoxic T-lymphocyte-associated antigen 4 receptor on T cells. It is associated with a range of immune-related toxicities. Sweets syndrome in association with ipilimumab has been reported only briefly in the literature. However, neutrophilic infiltration has been seen in biopsies of patients with ipilimumab-associated enterocolitis. We report, in detail, the case of a woman with metastatic melanoma undergoing ipilimumab therapy. After the second cycle of immunotherapy, the patient presented with high-grade fever followed by a rash on her hands. No infectious etiology was elucidated after an extensive workup. Pathologic examination of the skin biopsy from the hands confirmed neutrophilic dermatosis. The patient was treated with systemic steroids achieving complete remission of the skin lesions. Physicians should be aware of Sweets syndrome as a possible cutaneous side effect of ipilimumab therapy and be familiar with its management.


Journal of Clinical Oncology | 2017

Randomized, Open-Label Phase II Study Evaluating the Efficacy and Safety of Talimogene Laherparepvec in Combination With Ipilimumab Versus Ipilimumab Alone in Patients With Advanced, Unresectable Melanoma

Jason Chesney; Igor Puzanov; Frances A. Collichio; Parminder Singh; Mohammed M. Milhem; John A. Glaspy; Omid Hamid; Merrick I. Ross; Philip Friedlander; Claus Garbe; Theodore F. Logan; Axel Hauschild; Celeste Lebbe; Lisa Chen; Jenny J. Kim; Jennifer Gansert; Robert Hans Ingemar Andtbacka; Howard L. Kaufman

Purpose We evaluated the combination of talimogene laherparepvec plus ipilimumab versus ipilimumab alone in patients with advanced melanoma in a phase II study. To our knowledge, this was the first randomized trial to evaluate addition of an oncolytic virus to a checkpoint inhibitor. Methods Patients with unresectable stages IIIB to IV melanoma, with no more than one prior therapy if BRAF wild-type, no more than two prior therapies if BRAF mutant, measurable/injectable disease, and without symptomatic autoimmunity or clinically significant immunosuppression were randomly assigned 1:1 to receive talimogene laherparepvec plus ipilimumab or ipilimumab alone. Talimogene laherparepvec treatment began in week 1 (first dose, ≤ 4 mL × 106 plaque-forming units/mL; after 3 weeks, ≤ 4 mL × 108 plaque-forming units/mL every 2 weeks). Ipilimumab (3 mg/kg every 3 weeks; up to four doses) began week 1 in the ipilimumab alone arm and week 6 in the combination arm. The primary end point was objective response rate evaluated by investigators per immune-related response criteria. Results One hundred ninety-eight patients were randomly assigned to talimogene laherparepvec plus ipilimumab (n = 98), or ipilimumab alone (n = 100). Thirty-eight patients (39%) in the combination arm and 18 patients (18%) in the ipilimumab arm had an objective response (odds ratio, 2.9; 95% CI, 1.5 to 5.5; P = .002). Responses were not limited to injected lesions; visceral lesion decreases were observed in 52% of patients in the combination arm and 23% of patients in the ipilimumab arm. Frequently occurring adverse events (AEs) included fatigue (combination, 59%; ipilimumab alone, 42%), chills (combination, 53%; ipilimumab alone, 3%), and diarrhea (combination, 42%; ipilimumab alone, 35%). Incidence of grade ≥ 3 AEs was 45% and 35%, respectively. Three patients in the combination arm had fatal AEs; none were treatment related. Conclusion The study met its primary end point; the objective response rate was significantly higher with talimogene laherparepvec plus ipilimumab versus ipilimumab alone. These data indicate that the combination has greater antitumor activity without additional safety concerns versus ipilimumab.

Collaboration


Dive into the Philip Friedlander's collaboration.

Top Co-Authors

Avatar

Nina Bhardwaj

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Yvonne Saenger

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Lee Roy Morgan

Louisiana State University

View shared research outputs
Top Co-Authors

Avatar

Roy S. Weiner

University Medical Center New Orleans

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

William Oh

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Chrisann Kyi

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Ml Ware

Ochsner Medical Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge