J. A. Sosman
Loyola University Chicago
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by J. A. Sosman.
Journal of Clinical Oncology | 2005
David F. McDermott; Meredith M. Regan; Joseph I. Clark; Lawrence E. Flaherty; Geoffery R. Weiss; Theodore F. Logan; John M. Kirkwood; Michael S. Gordon; J. A. Sosman; Marc S. Ernstoff; Christopher P.G. Tretter; Walter J. Urba; John W. Smith; Kim Margolin; Jared A. Gollob; Janice P. Dutcher; Michael B. Atkins
PURPOSEnThe Cytokine Working Group conducted a randomized phase III trial to determine the value of outpatient interleukin-2 (IL-2) and interferon alfa-2b (IFN) relative to high-dose (HD) IL-2 in patients with metastatic renal cell carcinoma.nnnPATIENTS AND METHODSnPatients were stratified for bone and liver metastases, primary tumor in place, and Eastern Cooperative Oncology Group performance status 0 or 1 and then randomly assigned to receive either IL-2 (5 MIU/m(2) subcutaneously every 8 hours for three doses on day 1, then daily 5 days/wk for 4 weeks) and IFN (5 MIU/m(2) subcutaneously three times per week for 4 weeks) every 6 weeks or HD IL-2 (600,000 U/kg/dose intravenously every 8 hours on days 1 through 5 and 15 to 19 [maximum 28 doses]) every 12 weeks.nnnRESULTSnOne hundred ninety-two patients were enrolled between April 1997 and July 2000. Toxicities were as anticipated for these regimens. The response rate was 23.2% (22 of 95 patients) for HD IL-2 versus 9.9% (nine of 91 patients) for IL-2/IFN (P = .018). Ten patients receiving HD IL-2 were progression-free at 3 years versus three patients receiving IL-2 and IFN (P = .082). The median response durations were 24 and 15 [corrected] months (P = .18) [corrected] and median survivals were 17.5 and 13 months (P = .24). For patients with bone or liver metastases (P = .001) or a primary tumor in place (P = .040), survival was superior with HD IL-2.nnnCONCLUSIONnThis randomized phase III trial provides additional evidence that HD IL-2 should remain the preferred therapy for selected patients with metastatic renal cell carcinoma.
Journal of Clinical Oncology | 1993
J. A. Sosman; Geoffrey R. Weiss; Kim Margolin; Frederick R. Aronson; M Sznol; Michael B. Atkins; K R O'Boyle; Richard I. Fisher; D H Boldt; J Doroshow
PURPOSEnTo determine the maximum-tolerated dose (MTD) of an anti-CD3 antibody, OKT3, in combination with high-dose interleukin-2 (IL-2), and to determine whether OKT3 can enhance the expansion of CD3+, CD25+ (IL-2 receptor alpha [IL-2R alpha])-expressing T cells in the peripheral blood of patients with advanced melanoma and renal cell carcinoma receiving high-dose IL-2.nnnPATIENTS AND METHODSnWe performed a phase IB trial of a murine monoclonal anti-CD3 antibody (OKT3) with high-dose IL-2 in patients with advanced melanoma and renal cell carcinoma. Fifty-four patients were enrolled, with cohorts of 10 or more patients receiving escalating doses of OKT3 at 75, 200, 400, and 600 micrograms/m2 on day 1 followed by IL-2 at an initial dose 0.45 and then 1.33 mg/m2 every 8 hours on days 2 through 6 and 16 through 20 (maximum, 28 doses). An additional cohort of 14 patients received high-dose IL-2 (1.33 mg/m2 per dose) alone. Circulating CD3+, CD25+ cells were monitored before therapy and following the initial week of IL-2.nnnRESULTSnA total of 68 patients were enrolled. The MTD for OKT3 was defined as 400 micrograms/m2 based on a reduction in the number of IL-2 doses that could be administered. Increases in CD3+, CD25+ cells were observed within all cohorts; however, the increase was not OKT3 dose-dependent. On the other hand, we found that 60% (nine of 15) of patients tested at OKT3 dose levels of 200, 400, and 600 micrograms/m2 had increases in serum sCD25 (soluble IL-2R alpha) to more than 100,000 U/mL, while none of 10 patients who received IL-2 alone or with OKT3 at the 75-micrograms dose had increases greater than 60,000 U/mL. Of 29 patients with renal cell carcinoma who received OKT3 with IL-2 (1.33 mg/m2), there were three objective tumor responses (all partial responses). In the 16 patients with melanoma who received OKT3 plus IL-2, there was a single objective response (complete response).nnnCONCLUSIONnThe doses of OKT3 administered on this schedule failed to enhance significantly the number of circulating CD3+, CD25+ T cells and did not appear to increase the antitumor activity of IL-2 alone, which underscores the need for other approaches to enhance the efficacy of IL-2 therapy.
Journal of Immunotherapy | 1991
Gilda Weil-Hillman; Kathleen Schell; David M. Segal; Jacquelyn A. Hank; J. A. Sosman; Paul M. Sondel
Summary The role of activated T cells in the mediation of antitumor responses has been documented in several experimental models. In some of these, interleukin-2 (IL-2) has been used as a means to induce and expand the antitumor effects of the T cells. IL-2 has been tested in clinical trials for cancer treatment. Surprisingly, T cells appear to be inactivated by IL-2 in these clinical trials. T cells obtained from peripheral blood after IL-2 therapy showed decreased responses to mitogens and alloantigens, did not proliferate in vitro in response to IL-2, and did not mediate non-major histocompatibility complex-restricted cytotoxicity or targeted lysis in the presence of bispecific monoclonal antibodies. In this study, we present evidence that these post-IL-2 therapy T cells are not irreversibly inactivated; they can be activated in vitro by anti-CD3 monoclonal antibody together with IL-2 to upregulate the p55 component of the IL-2 receptor and proliferate. Nevertheless, following activation by anti-CD3 and IL-2, the level of targeted T-cell cytotoxicity mediated by the post-IL-2 therapy T cells was significantly lower than that by pre-IL-2 therapy T cells. Although in vivo treatment with IL-2 alone induces natural killer (NK) cells to mediate lymphokine-activated killer activity, these data suggest that the T-cell lytic function is inhibited by this treatment and only partially reversible by subsequent T-cell receptor activation using anti-CD3 mAb. Exposure of T cells to anti-CD3 mAb prior to in vivo IL-2 treatment generates T-cell lytic activity in vitro. These results, together with preclinical murine studies, suggest that a combined in vivo protocol of anti-CD3 mAb and IL-2, starting first with the anti-CD3 mAb, may cause activation of the T cells in addition to the activation of NK cells and thus warrant clinical testing.
Journal of Immunotherapy | 1995
Geoffrey R. Weiss; Kim Margolin; Mario Sznol; Michael B. Atkins; Leslie Oleksowicz; Randi Isaacs; J. A. Sosman; James H. Doroshow; Elizabeth Trehu; Janice P. Dutcher; Richard I. Fisher
IL-6 is a pluripotent cytokine with stimulatory effects on megakaryocytes, B lymphocytes, and (in combination with other cytokines) committed hematopoietic stem cells and T cells. IL-6 has direct antitumor activity against murine tumors. We previously completed a Phase I trial of 120-h continuous intravenous infusion of IL-6 repeated every 21 days and found 30 μg/kg/day to be the maximum tolerated dose (MTD), the dosage and schedule used for this Phase II trial. Eligibility requirements included histologic evidence of measurable advanced or metastatic renal cell carcinoma ; no prior immunotherapy for advanced disease ; performance status [PS ; Eastern Cooperative Oncology Group (ECOG)]≤1 ; and adequate hematologic, biochemical, and major organ function compatible with metabolism and safe tolerance of IL-6. Patients received IL-6 at a dosage of 30 μg/kg/day as a 120-h continuous infusion. Courses of therapy were repeated every 21 days. Patients were evaluated for response after every two courses of treatment. Fourteen patients were enrolled in this study : eight men and six women ; ages 34-75 years ; PS 0 :8 patients, PS 1 :6 patients. Twelve patients had prior nephrectomy. Thirty-five courses of IL-6 were administered. Two patients had partial responses of 6 and 8 months duration (14% response rate ; 95% CI : 2-45%). Eight patients had progressive disease, one patient had a minor response, and three patients had stable disease. Five patients developed atrial fibrillation occurring during the latter half of or soon after completion of the IL-6 infusion. One of these patients developed ischemic abnormalities on electrocardiogram, which resolved spontaneously without evidence of myocardial injury. One patient required electrical cardioversion. One patient required early suspension of treatment with elevation of serum bilirubin, and another developed moderate ataxia of gait. None of the patients experienced thrombocytosis during or after administration of IL-6. Although IL-6 can be administered on this schedule to patients, the modest antitumor activity and unacceptable toxicity of IL-6 of this schedule led to early termination of the study. However, this evidence of antitumor activity suggests that IL-6 should be studied on other schedules and perhaps in combination with other cytokines explored against renal cell carcinoma.
Journal of Immunotherapy | 1995
J. A. Sosman; Catherine Kefer; Richard I. Fisher; Cheryl D. Jacobs; Peggy Pumfery; Thomas M. Ellis
Summary: Preclinical studies have shown that anti-CD3 antibodies can enhance the in vitro activation of human T lymphocytes in combination with low-dose interleukin-2 (IL-2) and induce the in vivo rejection of murine tumors. A Phase IA/IB trial combining a murine monoclonal antibody, anti-CD3 antibody (OKT3), with low-dose continuous-infusion IL-2 was conducted in cancer patients to define the toxicity and immunologic effects of this combination. OKT3 administered weekly as a 15-min infusion at dose levels of 10, 100, 200, 400, and 600 üg/m2 was followed 18 h later by a 100-h infusion of IL-2 at 3 MIU/m2/day for 3 consecutive weeks. When feasible, patients also received the IL-2 course without OKT3 to assess the effects of OKT3 on the IL-2 regimen within the same patient. Thirty patients were enrolled onto the study, with 24 completing the OKT3/IL-2 course and 18 completing both OKT3/IL-2 and IL-2 alone courses. OKT3 administration was associated with acute hypotension with fevers of >40°C and in two patients cerebral vascular infarcts. At 600 üg/m2 OKT3, these toxicities were dose limiting. In a dose-dependent manner, OKT3 induced the transient release of tumor necrosis factor (TNF) and IL-6 into the serum and a profound lymphopenia. OKT3 did not significantly enhance the toxicity of this schedule of IL-2 administration. All solid tumor patients treated at 100–600 üg/m2 OKT3 showed induction of a human anti-murine antibody response prior to the third week of treatment. A patient with renal cell cancer treated at the 600-üg/m2 OKT3 dose level experienced a 4-month partial remission, and two mixed responses were observed in a sarcoma and a melanoma patient treated at 100− and 400-üg/m2 OKT3 dose levels, respectively. Most importantly, we were unable to demonstrate that the addition of OKT3 enhanced immune activation within peripheral blood based upon the magnitude of rebound lymphocytosis, increase in CD56+ or CD3+, CD25+ lymphocytes, induction of natural killer, lymphokine activated killer, or cytolytic T lymphocyte cytotoxicity, or release of serum cytokines (TNF, IL-6) or soluble CD25 (as assayed 24 h following IL-2 infusion). Therefore, this approach was ineffective at enhancing the immunologic effects of a low-dose continuous-infusion IL-2 regimen and will not be pursued further in clinical trials.
JCO Precision Oncology | 2017
Federica Catalanotti; Donavan T. Cheng; Alexander N. Shoushtari; Douglas B. Johnson; Katherine S. Panageas; Parisa Momtaz; Catherine Higham; Helen H. Won; James J. Harding; Taha Merghoub; Neal Rosen; J. A. Sosman; Michael F. Berger; Paul B. Chapman; David B. Solit
PurposenThe clinical use of BRAF inhibitors in patients with melanoma is limited by intrinsic and acquired resistance. We asked whether next-generation sequencing of pretreatment tumors could identify coaltered genes that predict for intrinsic resistance to BRAF inhibitor therapy in patients with melanoma as a prelude to rational combination strategies.nnnPatients and MethodsnWe analyzed 66 tumors from patients with metastatic BRAF-mutant melanoma collected before treatment with BRAF inhibitors. Tumors were analyzed for > 250 cancer-associated genes using a capture-based next-generation sequencing platform. Antitumor responses were correlated with clinical features and genomic profiles with the goal of identifying a molecular signature predictive of intrinsic resistance to RAF pathway inhibition.nnnResultsnAmong the 66 patients analyzed, 11 received a combination of BRAF and MEK inhibitors for the treatment of melanoma. Among the 55 patients treated with BRAF inhibitor monotherapy, objective responses, as assessed by Response Evaluation Criteria in Solid Tumors (RECIST), were observed in 30 patients (55%), with five (9%) achieving a complete response. We identified a significant association between alterations in PTEN that would be predicted to result in loss of function and reduced progression-free survival, overall survival, and response grade, a metric that combines tumor regression and duration of treatment response. Patients with melanoma who achieved an excellent response grade were more likely to have an elevated BRAF-mutant allele fraction.nnnConclusionnThese results provide a rationale for cotargeting BRAF and the PI3K/AKT pathway in patients with BRAF-mutant melanoma when tumors have concurrent loss-of-function mutations in PTEN. Future studies should explore whether gain of the mutant BRAF allele and/or loss of the wild-type allele is a predictive marker of BRAFi sensitivity.
JCO Precision Oncology | 2018
William Lu; Luciana Burton; James Larkin; Paul B. Chapman; Paolo Antonio Ascierto; A. Ribas; C. Robert; J. A. Sosman; Grant A. McArthur; Ilsung Chang; Ivor Caro; Elicia Penuel; Yibing Yan; Matthew Wongchenko
PurposeWe performed a retrospective exploratory analysis to evaluate the prognostic and predictive effect of two circulating biomarkers, BRAFV600 mutant circulating tumor DNA (ctDNA) and circulating hepatocyte growth factor (cHGF), in metastatic melanoma.Materials and MethodsThis study evaluated patients from BRIM-3, a phase III trial comparing vemurafenib and dacarbazine in 675 patients with BRAFV600 mutated advanced melanoma. ctDNA was measured using droplet digital polymerase chain reaction, and cHGF was measured by enzyme-linked immunosorbent assay. Overall survival (OS) was estimated using the Kaplan-Meier method, and hazard ratios (HRs) were estimated using Cox proportional hazards modeling. Partitioning analysis was used to group patients into risk categories.ResultsPatients with elevated levels of baseline BRAFV600 ctDNA had significantly shorter median OS than those with undetectable levels of ctDNA (vemurafenib arm, 9.9 v 21.4 months, respectively, and dacarbazine arm: 6.1 v 21.0 months, respect...
Journal of Clinical Oncology | 2009
Keith T. Flaherty; Igor Puzanov; J. A. Sosman; Kevin B. Kim; Antoni Ribas; Grant A. McArthur; Richard J. Lee; Joseph F. Grippo; Keith Nolop; Paul B. Chapman
Journal of Clinical Oncology | 2008
Reinhard Dummer; Caroline Robert; P. B. Chapman; J. A. Sosman; Mark R. Middleton; Lars Bastholt; K. Kemsley; Mireille Cantarini; C. Morris; John M. Kirkwood
Journal of Clinical Oncology | 2011
Antoni Ribas; Kevin B. Kim; Lynn Schuchter; Rene Gonzalez; Anna C. Pavlick; Jeffrey S. Weber; Grant A. McArthur; Thomas E. Hutson; Keith T. Flaherty; Stergios J. Moschos; D. P. Lawrence; Peter Hersey; Richard F. Kefford; Bartosz Chmielowski; Igor Puzanov; Jiang Li; Keith Nolop; Rebecca Lee; Andrew K. Joe; J. A. Sosman