Network


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

Hotspot


Dive into the research topics where Philip Schulman is active.

Publication


Featured researches published by Philip Schulman.


The New England Journal of Medicine | 1994

Intensive Postremission Chemotherapy in Adults with Acute Myeloid Leukemia

Robert J. Mayer; Roger B. Davis; Charles A. Schiffer; Deborah T. Berg; Bayard L. Powell; Philip Schulman; George A. Omura; Joseph O. Moore; Or McIntyre; Emil Frei

BACKGROUND About 65 percent of previously untreated adults with primary acute myeloid leukemia (AML) enter complete remission when treated with cytarabine and an anthracycline. However, such responses are rarely durable when conventional postremission therapy is administered. Uncontrolled trials have suggested that intensive postremission therapy may prolong these complete remissions. METHODS We treated 1088 adults with newly diagnosed AML with three days of daunorubicin and seven days of cytarabine and randomly assigned patients who had a complete remission to receive four courses of cytarabine at one of three doses: 100 mg per square meter of body-surface area per day for five days by continuous infusion, 400 mg per square meter per day for five days by continuous infusion, or 3 g per square meter in a 3-hour infusion every 12 hours (twice daily) on days 1, 3, and 5. All patients then received four courses of monthly maintenance treatment. RESULTS Of the 693 patients who had a complete remission, 596 were randomly assigned to receive postremission cytarabine. After a median follow-up of 52 months, the disease-free survival rates in the three treatment groups were significantly different (P = 0.003). Relative to the 100-mg group, the hazard ratios were 0.67 for the 3-g group (95 percent confidence interval, 0.53 to 0.86) and 0.75 for the 400-mg group (95 percent confidence interval, 0.60 to 0.94). The probability of remaining in continuous complete remission after four years for patients 60 years of age or younger was 24 percent in the 100-mg group, 29 percent in the 400-mg group, and 44 percent in the 3-g group (P = 0.002). In contrast, for patients older than 60, the probability of remaining disease-free after four years was 16 percent or less in each of the three postremission cytarabine groups. CONCLUSIONS These data support the concept of a dose-response effect for cytarabine in patients with AML who are 60 years of age or younger. The results with the high-dose schedule in this age group are comparable to those reported in similar patients who have undergone allogeneic bone marrow transplantation during a first remission.


The New England Journal of Medicine | 1995

Granulocyte–Macrophage Colony-Stimulating Factor after Initial Chemotherapy for Elderly Patients with Primary Acute Myelogenous Leukemia

Richard Stone; Deborah T. Berg; Stephen L. George; Richard K. Dodge; Paolo Alberto Paciucci; Philip Schulman; Edward J. Lee; Joseph O. Moore; Bayard L. Powell; Charles A. Schiffer

Background Elderly patients with primary acute myelogenous leukemia (AML) are less likely to enter remission than younger adults, in part because of a higher mortality rate related to severe myelosuppression. Granulocyte–macrophage colony-stimulating factor (GM-CSF) has been shown to shorten the duration of neutropenia and decrease infectious complications when administered after chemotherapy to patients with lymphomas and solid tumors. Methods We randomly assigned 388 patients 60 years of age or older who had newly diagnosed primary AML to receive placebo or GM-CSF (5 μg per kilogram of body weight per day intravenously) in a double-blind manner, beginning the day after the completion of three days of daunorubicin and seven days of cytarabine. If leukemic cells persisted in the marrow three weeks after the initiation of chemotherapy, further daunorubicin (two days) and cytarabine (five days) were administered. GM-CSF or placebo was given daily until the neutrophil count was at least 1000 per cubic millim...


Journal of Clinical Oncology | 2005

Phase III Randomized Multicenter Study of a Humanized Anti-CD33 Monoclonal Antibody, Lintuzumab, in Combination With Chemotherapy, Versus Chemotherapy Alone in Patients With Refractory or First-Relapsed Acute Myeloid Leukemia

Eric J. Feldman; Joseph Brandwein; Richard Stone; Matt Kalaycio; Joseph O. Moore; Julie O'Connor; Nancy Wedel; Gail J. Roboz; Carole B. Miller; Rajesh Chopra; Joseph C Jurcic; Randy A. Brown; W. Christopher Ehmann; Philip Schulman; Stanley R. Frankel; Daniel J. De Angelo; David A. Scheinberg

PURPOSE Lintuzumab (HuM195) is an unconjugated humanized murine monoclonal antibody directed against the cell surface myelomonocytic differentiation antigen CD33. In this study, the efficacy of lintuzumab in combination with induction chemotherapy was compared with chemotherapy alone in adults with first relapsed or primary refractory acute myeloid leukemia (AML). PATIENTS AND METHODS Patients with relapsed or primary resistant AML (duration of first response, zero to 12 months) were randomly assigned to receive either mitoxantrone 8 mg/m(2), etoposide 80 mg/m(2), and cytarabine 1 g/m(2) daily for 6 days (MEC) in combination with lintuzumab 12 mg/m(2), or MEC alone. Overall response, defined as the rate of complete remission (CR) and CR with incomplete platelet recovery (CRp), was the primary end point of the study, with additional analyses of survival time and toxicity. RESULTS A total of 191 patients were randomly assigned from November 1999 to April 2001. The percent CR plus CRp with MEC plus lintuzumab was 36% v 28% in patients treated with MEC alone (P = .28). The overall median survival was 156 days and was not different in the two arms of the study. Apart from mild antibody infusion-related toxicities (fever, chills, and hypotension), no differences in chemotherapy-related adverse effects, including hepatic and cardiac dysfunction, were observed with the addition of lintuzumab to induction chemotherapy. CONCLUSION The addition of lintuzumab to salvage induction chemotherapy was safe, but did not result in a statistically significant improvement in response rate or survival in patients with refractory/relapsed AML.


Leukemia | 2003

Treatment of relapsed or refractory acute myeloid leukemia with humanized anti-CD33 monoclonal antibody HuM195.

Feldman Ej; M Kalaycio; G. Weiner; S. Frankel; Philip Schulman; L. Schwartzberg; J. Jurcic; E. Velez-Garcia; Karen Seiter; David A. Scheinberg; D. Levitt; N. Wedel

HuM195 is a humanized, unconjugated, anti-CD33 monoclonal antibody. Fifty adult patients with relapsed or refractory AML were randomized to receive HuM195 at a dose of 12 or 36 mg/m2 by intravenous infusion over 4 h on days 1–4 and 15–18. Patients with stable or responding disease received two additional cycles on days 29–32 and 43–46. HuM195 was given as first salvage therapy in 24 patients and as second or subsequent salvage therapy in 26 patients. Pretreatment blast percentage in the marrow was between 5 and 30% in 20 patients with the others having blast counts greater than 30%. The median age of patients was 62 years (range 26–86) and CD33 was detected in 95% of patients for whom immunophenotyping was available. Of 49 evaluable patients, two complete and one partial remission were observed. All three responses were in patients treated at the 12 mg/m2 dose level and all had baseline blast percentages less than 30%. Decreases in blast counts ranging from 30 to 74% were seen in nine additional patients. Infusion-related events of fever and chills occurred in the majority of patients and were generally mild and primarily related to the first dose of antibody. No hepatic, renal or cardiac toxicities were observed and other adverse events such as nausea, vomiting, mucositis and diarrhea were uncommon or felt to be unrelated to HuM195. In addition, anti-HuM195 responses were not detected. HuM195 as a single agent has minimal, but observable, anti-leukemic activity in patients with relapsed or refractory AML and activity is confined to patients with low burden disease. No significant differences in clinical efficacy or toxicity were seen between the two dose levels of antibody. HuM195 was well tolerated with infusion-related fevers and chills the predominant toxicities seen. Meaningful clinical efficacy of this unconjugated monoclonal antibody may be realized only in patients with minimal residual disease, or in combination with chemotherapy.


Journal of Clinical Investigation | 1996

Examples of in vivo isotype class switching in IgM+ chronic lymphocytic leukemia B cells.

Franco Fais; Brian Sellars; Fabio Ghiotto; Xiao Jie Yan; Mariella Dono; Steven L. Allen; Daniel R. Budman; Klaus Dittmar; Jonathan E. Kolitz; Stuart M. Lichtman; Philip Schulman; Michael W. Schuster; Vincent Vinciguerra; Kanti R. Rai; Freda K. Stevenson; Peter K. Gregersen; Manlio Ferrarini; Nicholas Chiorazzi

Chronic lymphocytic leukemia (CLL) usually involves the expansion of a clone of CD5+ B cells synthesizing IgM antibodies. These B cells appear to be blocked at the antigen receptor-expressing stage of B cell differentiation and are thought not to undergo an isotype class switch to IgG or IgA production. In vivo and in vitro studies suggest, however, that in some instances terminal differentiation and isotype switching can occur. To test the hypothesis that in vivo isotype class switching occurs in IgM+ B-type CLL cells, we analyzed the PBMC of 19 CLL patients for the presence of transcripts encoding the rearranged CLL V(H)DJ(H) associated with either gamma or alpha H chains. The molecular data indicate that approximately 50% of B-CLL patients have amplifications of IgM+ B cells that undergo an isotype class switch. Switching to IgA appears to occur more often than to IgG; also, switching can involve different IgG subclasses in individual patients. In many instances, these CLL-related gamma and alpha transcripts are much more plentiful than those of normal B cells that produce the same isotype. These switched transcripts do not reveal evidence for the accumulation of significant numbers of new V(H) gene mutations. The cellular data indicate that B cells with lesser amounts of surface membrane IgD and higher IgM/IgD ratios are more likely to undergo this switching process. Furthermore, B cells expressing IgG and IgA of the same idiotype or V(H) family and the same CDR3 length as those of the CLL IgM+ clone can be identified in the blood of patients studied using multiparameter immunofluorescence analyses. Collectively, these data suggest that not all members of a B-CLL clone are frozen at the surface membrane Ig-expressing stage of B cell maturation, and that some members can switch to the production of non-IgM isotypes. The occurrence of switching without the accumulation of V gene mutations indicates that the processes of differentiation and diversification are not linked.


Journal of Clinical Oncology | 1994

Phase I evaluation of a water-soluble etoposide prodrug, etoposide phosphate, given as a 5-minute infusion on days 1, 3, and 5 in patients with solid tumors.

Daniel R. Budman; L N Igwemezie; S Kaul; J Behr; Stuart M. Lichtman; Philip Schulman; Vincent Vinciguerra; Steven L. Allen; Jonathan E. Kolitz; K Hock

PURPOSE To determine the toxicities, maximum-tolerated dose (MTD), and pharmacology of etoposide phosphate, a water-soluble etoposide derivative, administered as a 5-minute intravenous infusion on a schedule of days 1, 3, and 5 repeated every 21 days. PATIENTS AND METHODS Thirty-six solid tumor patients with a mean age of 63 years, performance status of 0 to 1, WBC count > or = 4,000/microL, and platelet count > or = 100,000/microL, with normal hepatic and renal function were studied. Doses evaluated in etoposide equivalents were 50, 75, 100, 125, 150, 175, and 200 mg/m2/d. Etoposide in plasma and urine and etoposide phosphate in plasma were measured by high-performance liquid chromatography (HPLC). Eleven of 36 patients were treated with concentrated etoposide phosphate at 150 mg/m2/d. RESULTS Grade I/II nausea, vomiting, alopecia, and fatigue were common. Leukopenia (mainly neutropenia) occurred at doses greater than 75 mg/m2, with the nadir occurring between days 15 and 19 posttreatment. All effects were reversible. Hypotension, bronchospasm, and allergic reactions were not observed in the first 25 patients. The MTD due to leukopenia was determined to be between 175 and 200 mg/m2/d. In 11 patients treated with concentrated etoposide phosphate, no local phlebitis was noted, but two patients did develop allergic phenomena. The conversion of etoposide phosphate to etoposide was not saturated in the dosages studied. Etoposide phosphate had peak plasma concentrations at 5 minutes, with a terminal half-life (t1/2) of 7 minutes. Etoposide reached peak concentrations at 7 to 8 minutes, with a t1/2 of 6 to 9 hours. Both etoposide phosphate and etoposide demonstrated dose-related linear increases in maximum plasma concentration (Cmax) and area under the curve (AUC). CONCLUSION Etoposide phosphate displays excellent patient tolerance in conventional dosages when administered as a 5-minute intravenous bolus. The suggested phase II dose is 150 mg/m2 on days 1, 3, and 5. The ability to administer etoposide phosphate as a concentrated, rapid infusion may prove of value both in the outpatient clinic and in high-dose regimens.


Cancer | 1982

Cytogenetic findings in the dysmyelopoietic syndrome.

Nataline Kardon; Philip Schulman; Thomas J. Degnan; Daniel R. Budman; Jessica G. Davis; Vincent Vinciguerra

Cytogenetic studies of bone marrow specimens from 15 patients with dysmyelopoietic syndrome are presented. The group consists of nine patients with refractory anemia with excess of blasts (RAEB), three patients with chronic myelomonocytic leukemia (CMMoL), and three patients with acquired idiopathic sideroblastic anemia (AISA). None of these patients had a prior history of therapeutic or occupational exposure to potential carcinogenic agents. G(TG)‐banding revealed clonal abnormalities in nine of the 15 patients. Five of these patients exhibited one or more of the following cytogenetic abnormalities: 5q deletion, −7, +8, or +21. The AISA group appeared to be unique as chromosome abnormalities were seen in two of the three patients and the clinical course in these patients has been prolonged without progression to acute leukemia. No other clinical correlations could be made in the blast RAEB and CMMoL groups, except for possible survival benefit in patients with normal karyotypes.


Leukemia Research | 2011

High Dose Cytarabine plus Gemtuzumab Ozogamicin for Patients with Relapsed or Refractory Acute Myeloid Leukemia: Cancer and Leukemia Group B Study 19902

Richard Stone; Barry K. Moser; Ben Sanford; Philip Schulman; Jonathan E. Kolitz; Steven L. Allen; Wendy Stock; Ilene Galinsky; Ravi Vij; Guido Marcucci; David D. Hurd; Richard A. Larson

Gemtuzumab ozogamicin (GO), an anti-CD33 immunoconjugate, was combined with high dose cytarabine (HiDAC; cytarabine 3g/m(2) over 3h daily for 5 days) for adults with relapsed or refractory AML. HiDAC plus GO 9mg/m(2) on day 7 and 4.5mg/m(2) on day 14 was not tolerated, but HiDAC followed by GO 9mg/m(2) on day 7 was safe: 12/37 (32%) patients with relapsed AML achieved complete remission. Median overall survival was 8.9 months. No grade 4 hepatic veno-occlusive disease was observed. This regimen merits further study, both in this setting and as a remission consolidation therapy.


Clinical Cancer Research | 2011

Phase I Trial of Weekly and Twice-Weekly Bortezomib with Rituximab, Cyclophosphamide, and Prednisone in Relapsed or Refractory Non-Hodgkin Lymphoma

John F. Gerecitano; Carol S. Portlock; Paul A. Hamlin; Craig H. Moskowitz; Ariela Noy; David Straus; Philip Schulman; Otilia Dumitrescu; Debra Sarasohn; Jennifer Pappanicholaou; Alexia Iasonos; Zhigang Zhang; Qianxing Mo; Endri Horanlli; Celeste Rojas; Andrew D. Zelenetz; Owen A. O'Connor

Purpose: To determine the safety and efficacy of substituting weekly or twice-weekly bortezomib for vincristine in the R-CVP (rituximab, cyclophosphamide, vincristine, and prednisone) regimen in patients with relapsed/refractory indolent and mantle cell lymphoma (MCL). Experimental Design: Of the 57 patients in this phase I trial, 55 participated in 1 of 2 dosing schedules that included rituximab (375 mg/m2) and cyclophosphamide (750 or 1,000 mg/m2) administered on day 1 of each 21-day cycle and prednisone (100 mg orally) days 2 to 6. In the once-weekly schedule, bortezomib was administered on days 2 and 8; on the twice-weekly schedule, bortezomib was given on days 2, 5, 9, and 12. Bortezomib and cyclophosphamide were alternately escalated. A separate cohort of 10 patients in the twice-weekly schedule received concurrent pegfilgrastim (PegG) on day 2. Results: Both schedules of R-CBorP (rituximab, cyclophosphamide, bortezomib, and prednisone) were well tolerated. Most toxicities across all dose levels and cycles were grade 1 or 2. The overall response rates for patients on the weekly (n = 13) and twice-weekly (n = 33) schedules were 46% [23% complete response/complete response unconfirmed (CR/CRu)] and 64% (36% CR/CRu), respectively. Concurrent PegG did not increase hematologic toxicities in this regimen. A randomized phase II study is under way to further compare toxicity and efficacy of the 2 dosing schedules. Conclusions: R-CBorP is a safe and effective regimen in patients with relapsed/refractory indolent and MCLs. Most toxicities were grade 1 or 2, and a promising response rate was seen in this phase I study. Clin Cancer Res; 17(8); 2493–501. ©2011 AACR.


British Journal of Haematology | 1990

Deferoxamine-induced restoration of haematopoiesis in myelofibrosis secondary to myelodysplasia.

Jonathan H. Marsh; Michael Hundert; Philip Schulman

Primary myelofibrosis (MF), a disorder characterized by the abnormal deposition of collagen in the bone marrow without any recognizable underlying neoplastic disorder, has been well described (Kelsey & Geary, 1987; Smith et al, 1988). Secondary MF can occur in the setting of a myeloproliferative or any other neoplastic disorder. Deferoxamine is a chelating agent used to treat states of iron excess. We report a case of MF. treated supportively with transfusions for almost 7 years with an average transfusion requirement of 2 units/month, and pancytopenia, who had restoration of active haematopoiesis and ablation of his transfusion requirement following initiation of deferoxamine therapy. F.B. is a 79-year-old white male who presented in 1982 with progressive pancytopenia with no evidence of hepatosplenomegaly. Initial laboratory measurements were: WBC 1-6 x 10y/l: Hb 9.5 g/dl; PCV 29.3%: MCV 92 fl platelet count 45 x l O y / 1 and lactate dehydrogenase (LDH) 93 5 IU/I. Peripheral smear revealed a marked decrease in platelets and WBC, normochromic normocytic RBC with moderate numbers of schistocytes and rare teardrops. Bone marrow aspirate revealed erythroid hyperplasia with moderate dyserythropoiesis and a shift to the left in the myeloid series; the biopsy was hypercellular with increased collagen. Further workup was unremarkable. Cytogenetic studies revealed trisomy 8 in 19 of 20 metaphases analysed. The patient was felt to have MF in the setting of a dysmyelopoietic syndrome versus primary MF. The patient was subsequently treated at various times with anadrol (oxymetholene) up to 200 mg daily with or without pyridoxine, and prednisone up to 20 mg daily. His transfusion requirements varied from 1 unit of packed red blood cells every 3 weeks to 3 units, packed red blood cells every month. Other than mild progressive splenomegaly, his condition remained unchanged. A repeat bone marrow in 19 84 revealed marked hypercellularity with atypical megakaryocytes and increased stromal reticulum. In 1988 he was started on diuretic therapy for mild congestive heart failure, and was noted to have a ferritin of 5430 &I. In December 1988 he started deferoxamine 2000 mg subcutaneously over 8 h daily (30 mg/kg/d). Over the next 8 months he became transfusion free. His ferritin was initially noted to increase (8630 &I2 months into therapy), but then started to decrease (6507 pg/l in July 1989). His mild splenomegaly persisted, and his most recent bone marrow examination in July 1989 revealed a hypercellular biopsy with myeloid hyperplasia, atypical megakaryocytosis and fibrosis, consistent with a myeloproliferative or myelodysplastic disorder. This was not significantly different from his earlier biopsies. It should be noted that no pathological sideroblasts were observed in this marrow, nor in any of his prior marrows. The blood count at that time revealed: WBC 7.2 x 1OY//1 (74% polymorphs, 11% band cells, 8% lymphocytes, 3% basophils, 2% monocytes and 2% nucleated red blood cells), Hb 11.1 g/dl, PCV 34.8% and platelet count

Collaboration


Dive into the Philip Schulman's collaboration.

Top Co-Authors

Avatar

Vincent Vinciguerra

North Shore University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Stuart M. Lichtman

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kanti R. Rai

North Shore-LIJ Health System

View shared research outputs
Top Co-Authors

Avatar

Thomas J. Degnan

North Shore University Hospital

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge