Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 65
1.
Ann Oncol ; 33(2): 204-215, 2022 02.
Article En | MEDLINE | ID: mdl-34710571

BACKGROUND: Antitumor activity of ipilimumab or BRAF ± MEK inhibitors (BRAFi ± MEKi) following pembrolizumab administration in melanoma is poorly characterized. PATIENTS AND METHODS: In the phase III KEYNOTE-006 study, patients with unresectable stage III/IV melanoma received pembrolizumab (10 mg/kg) once every 2 or 3 weeks (Q3W) or ipilimumab (3 mg/kg) Q3W. The current post hoc analysis evaluates outcomes with ipilimumab or BRAFi ± MEKi as first subsequent systemic therapy after pembrolizumab administration and includes patients who completed or discontinued pembrolizumab after one or more dose. Pembrolizumab arms were pooled. RESULTS: At data cut-off (4 December 2017), median follow-up was 46.9 months. Of 555 pembrolizumab-treated patients, first subsequent therapy was ipilimumab for 103 (18.6%) and BRAFi ± MEKi for 59 (10.6%) [33 received BRAFi + MEKi, 26 BRAFi alone; 37 (62.7%) were BRAFi ± MEKi naïve]. In the subsequent ipilimumab group, ORR with previous pembrolizumab was 17.5% [1 complete response (CR); 17 partial response (PR)]; 79.6% had discontinued pembrolizumab due to progressive disease (PD); median overall survival (OS) was 21.5 months. ORR with subsequent ipilimumab was 15.5%; 11/16 responses (8 CRs; 3 PRs) were ongoing. ORR with subsequent ipilimumab was 9.7% for patients with PD as best response to pembrolizumab. Median OS from ipilimumab initiation was 9.8 months. In the subsequent BRAFi ± MEKi group, ORR with previous pembrolizumab was 13.5% (8 PR); 76.3% had discontinued pembrolizumab due to PD; median OS was 17.9 months. ORR with subsequent BRAFi ± MEKi was 30.5%, 7/18 responses (4 CR, 3 PR) were ongoing. Median OS from BRAFi ± MEKi initiation was 12.9 months. ORR for BRAFi ± MEKi-naïve patients who received subsequent BRAFi ± MEKi was 43.2%; 6/16 were ongoing (3 CR, 3 PR). CONCLUSIONS: Ipilimumab and BRAFi ± MEKi have antitumor activity as first subsequent therapy after pembrolizumab in patients with advanced melanoma.


Melanoma , Proto-Oncogene Proteins B-raf , Antibodies, Monoclonal, Humanized , Humans , Ipilimumab/adverse effects , Melanoma/pathology , Mitogen-Activated Protein Kinase Kinases/therapeutic use
3.
Ann Oncol ; 28(8): 1988-1995, 2017 Aug 01.
Article En | MEDLINE | ID: mdl-28595336

BACKGROUND: Surrogate biomarkers of efficacy are needed for anti-PD1/PD-L1 therapy, given the existence of delayed responses and pseudo-progressions. We evaluated changes in serum IL-8 levels as a biomarker of response to anti-PD-1 blockade in melanoma and non-small-cell lung cancer (NSCLC) patients. PATIENTS AND METHODS: Metastatic melanoma and NSCLC patients treated with nivolumab or pembrolizumab alone or nivolumab plus ipilimumab were studied. Serum was collected at baseline; at 2-4 weeks after the first dose; and at the time-points of response evaluation. Serum IL-8 levels were determined by sandwich ELISA. Changes in serum IL-8 levels were compared with the Wilcoxon test and their strength of association with response was assessed with the Mann-Whitney test. Accuracy of changes in IL-8 levels to predict response was estimated using receiver operation characteristics curves. RESULTS: Twenty-nine melanoma patients treated with nivolumab or pembrolizumab were studied. In responding patients, serum IL-8 levels significantly decreased between baseline and best response (P <0.001), and significantly increased upon progression (P =  0.004). In non-responders, IL-8 levels significantly increased between baseline and progression (P =  0.013). Early changes in serum IL-8 levels (2-4 weeks after treatment initiation) were strongly associated with response (P <0.001). These observations were validated in 19 NSCLC patients treated with nivolumab or pembrolizumab (P =  0.001), and in 15 melanoma patients treated with nivolumab plus ipilimumab (P <0.001). Early decreases in serum IL-8 levels were associated with longer overall survival in melanoma (P =  0.001) and NSCLC (P =  0.015) patients. Serum IL-8 levels also correctly reflected true response in three cancer patients presenting pseudoprogression. CONCLUSIONS: Changes in serum IL-8 levels could be used to monitor and predict clinical benefit from immune checkpoint blockade in melanoma and NSCLC patients.


Antineoplastic Agents, Immunological/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , Carcinoma, Non-Small-Cell Lung/drug therapy , Interleukin-8/blood , Lung Neoplasms/drug therapy , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Adult , Antineoplastic Agents, Immunological/pharmacology , Carcinoma, Non-Small-Cell Lung/blood , Cohort Studies , Enzyme-Linked Immunosorbent Assay , Female , Humans , Lung Neoplasms/blood , Male , Melanoma/blood , Middle Aged , Skin Neoplasms/blood , Survival Analysis
4.
Oncogene ; 30(13): 1542-50, 2011 Mar 31.
Article En | MEDLINE | ID: mdl-21119596

MicroRNAs (miRNAs) are small ∼22nt single stranded RNAs that negatively regulate protein expression by binding to partially complementary sequences in the 3' untranslated region (3' UTRs) of target gene messenger RNAs (mRNA). Recently, mutations have been identified in both miRNAs and target genes that disrupt regulatory relationships, contribute to oncogenesis and serve as biomarkers for cancer risk. KIT, an established oncogene with a multifaceted role in melanogenesis and melanoma pathogenesis, has recently been shown to be upregulated in some melanomas, and is also a target of the miRNA miR-221. Here, we describe a genetic variant in the 3' UTR of the KIT oncogene that correlates with a greater than fourfold increased risk of acral melanoma. This KIT variant results in a mismatch in the seed region of a miR-221 complementary site and reporter data suggests that this mismatch can result in increased expression of the KIT oncogene. Consistent with the hypothesis that this is a functional variant, KIT mRNA and protein levels are both increased in the majority of samples harboring the KIT variant. This work identifies a novel genetic marker for increased heritable risk of melanoma.


3' Untranslated Regions/genetics , Melanoma/genetics , MicroRNAs/physiology , Oncogenes , Proto-Oncogene Proteins c-kit/genetics , Skin Neoplasms/genetics , Case-Control Studies , Humans , Melanoma/etiology , Protein Biosynthesis , RNA, Messenger/analysis , Risk , Skin Neoplasms/etiology
5.
Blood ; 97(7): 1942-6, 2001 Apr 01.
Article En | MEDLINE | ID: mdl-11264156

We conducted a phase II randomized trial of recombinant granculocyte-macrophage colony-stimulating factor (GM-CSF) administered before topotecan chemotherapy to determine whether it could prevent myelosuppression and to determine the antitumor activity of this topoisomerase I inhibitor in 53 patients with metastatic malignant melanoma and renal cell cancer. All patients received GM-CSF after topotecan at a dose of 250 microg/m(2) daily for at least 8 days. Patients randomly assigned to receive GM-CSF priming were treated with GM-CSF at 250 microg/m(2) twice daily for 5 days before treatment. Twenty-five patients were randomly assigned to receive GM-CSF priming and 28 to receive topotecan without priming. The primary analysis was restricted to the protective effects seen during the first cycle of therapy. Grade 4 neutropenia occurred in 8 of 23 patients (35%) and grade 3 neutropenia in 5 of 23 patients (22%) randomized to GM-CSF priming, whereas 18 of 26 (69%) and 5 of 26 (19%) patients experienced grade 4 or 3 neutropenia, respectively, without GM-CSF priming (P =.0074). The mean duration of neutropenia was reduced by GM-CSF priming: grade 3 neutropenia from 5.2 +/- 0.7 to 2.8 +/- 0.7 days (P =.0232) and grade 4 neutropenia from 2.7 +/- 0.6 to 1.1 +/- 0.4 days (P = 0.0332). The protective effects of GM-CSF extended to the second cycle of treatment. The incidence of febrile neutropenia was also reduced. Chemotherapy-induced anemia and thrombocytopenia were similar in both groups. One partial response was seen in a patient with melanoma, and one patient with renal cell cancer had complete regression of pulmonary metastases and was rendered disease-free by nephrectomy. (Blood. 2001;97:1942-1946)


Antineoplastic Agents/adverse effects , Carcinoma, Renal Cell/drug therapy , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Kidney Neoplasms/drug therapy , Melanoma/drug therapy , Neutropenia/prevention & control , Skin Neoplasms/drug therapy , Topotecan/adverse effects , Anemia/chemically induced , Antineoplastic Agents/therapeutic use , Carcinoma, Renal Cell/blood , Diabetes Mellitus, Type 1/complications , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Granulocyte-Macrophage Colony-Stimulating Factor/adverse effects , Humans , Hypotension/chemically induced , Kidney Neoplasms/blood , Melanoma/blood , Neutropenia/chemically induced , Premedication , Prospective Studies , Remission Induction , Severity of Illness Index , Skin Neoplasms/blood , Stroke/etiology , Thrombocytopenia/chemically induced , Topotecan/therapeutic use , Treatment Outcome
10.
Cancer J Sci Am ; 6 Suppl 1: S11-4, 2000 Feb.
Article En | MEDLINE | ID: mdl-10685652

PURPOSE: To update response duration and survival data for patients with metastatic melanoma receiving the high-dose IV bolus recombinant interleukin (IL)-2 regimen. PATIENTS AND METHODS: Two hundred seventy assessable patients were entered into eight clinical trials conducted between 1985 and 1993. IL-2 600,000 or 720,000 IU/kg was administered by 15-minute intravenous infusion every 8 hours for up to 14 consecutive doses over 5 days as clinically tolerated with maximum support, including pressors. A second, identical cycle of treatment was scheduled following 6 to 9 days of rest, and courses could be repeated every 6 to 12 weeks in stable or responding patients. Responding patients received up to five courses (two cycles/course) of treatment. All data were updated through December 1998 using report forms completed by the clinical investigators. RESULTS: The objective overall response rate was unchanged from the previous report. Tumor responses were seen in 16% of patients, with complete responses in 17 (6%) and partial responses in 26 (10%). Median survival for the group as a whole is now 12 months. Median follow-up time for surviving patients exceeds 7 years. Median duration of response for the 43 responding patients and the 26 patients with partial responses remained unchanged at 8.9 and 5.9 months, respectively. Response durations ranged from 1.5 to > 122 months. The median duration of complete responses has yet to be reached, but is at least 59 months. Thirty-one patients (11%) were alive as of last contact; 28 were confirmed, including 18 responding patients. Three patients were lost to follow-up at > 1, > 13, and > 104 months. Twelve responding patients remained continually disease- or progression-free from > 70 to > 150 months following initiation of therapy. Disease progression was not observed in any patient who was responding as of the last report or in any patient responding for longer than 30 months. CONCLUSION: These data continue to support the notion that high-dose IL-2 produces durable responses in some patients with metastatic melanoma and should be considered a therapeutic option for appropriately selected patients with this disease.


Interleukin-2/therapeutic use , Melanoma/therapy , Follow-Up Studies , Humans , Melanoma/mortality , Melanoma/secondary , Recombinant Proteins/therapeutic use , Survival Rate
11.
J Clin Oncol ; 17(7): 2105-16, 1999 Jul.
Article En | MEDLINE | ID: mdl-10561265

PURPOSE: To determine the short- and long-term efficacy and toxicity of the high-dose intravenous bolus interleukin 2 (IL-2) regimen in patients with metastatic melanoma. PATIENTS AND METHODS: Two hundred seventy assessable patients were entered onto eight clinical trials conducted between 1985 and 1993. IL-2 (Proleukin [aldesleukin]; Chiron Corp, Emeryville, CA) 600,000 or 720,000 IU/kg was administered by 15-minute intravenous infusion every 8 hours for up to 14 consecutive doses over 5 days as clinically tolerated with maximum support, including pressors. A second identical treatment cycle was scheduled after 6 to 9 days of rest, and courses could be repeated every 6 to 12 weeks in stable or responding patients. Data were analyzed through fall 1996. RESULTS: The overall objective response rate was 16% (95% confidence interval, 12% to 21%); there were 17 complete responses (CRs) (6%) and 26 partial responses (PRs) (10%). Responses occurred with all sites of disease and in patients with large tumor burdens. The median response duration for patients who achieved a CR has not been reached and was 5.9 months for those who achieved a PR. Twelve (28%) of the responding patients, including 10 (59%) of the patients who achieved a CR, remain progression-free. Disease did not progress in any patient responding for more than 30 months. Baseline performance status and whether patients had received prior systemic therapy were the only predictive prognostic factors for response to IL-2 therapy. Toxicities, although severe, generally reversed rapidly after therapy was completed. Six patients (2%) died from adverse events, all related to sepsis. CONCLUSION: High-dose IL-2 treatment seems to benefit some patients with metastatic melanoma by producing durable CRs or PRs and should be considered for appropriately selected melanoma patients.


Interleukin-2/administration & dosage , Melanoma/drug therapy , Melanoma/secondary , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Disease-Free Survival , Female , Humans , Interleukin-2/adverse effects , Male , Melanoma/mortality , Middle Aged , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects , Risk Factors , Salvage Therapy , Survival Rate , Treatment Outcome , United States/epidemiology
12.
J Immunol ; 163(3): 1690-5, 1999 Aug 01.
Article En | MEDLINE | ID: mdl-10415076

Patients with metastatic melanoma were immunized with an immunodominant peptide derived from the gp100 melanoma-melanocyte differentiation Ag that was modified to increase binding to HLA-A+0201. A total of 10 of 11 patients who received the g209-2M peptide alone developed precursors reactive with the native g209 peptide, compared with only 5 of 16 patients who received g209-2M peptide plus IL-2 (p2 = 0.005). Peptide reactivity closely correlated with the recognition of HLA-A+0201 melanoma cells (p < 0. 001). The decrease in immune reactivity when peptide was administered with IL-2 appeared specific for the immunizing peptide, since reactivity to an influenza peptide resulting from prior exposure was not affected. Preexisting antitumor precursors did not decrease when peptide plus IL-2 was administered. The administration of GM-CSF or IL-12 also resulted in a decrease in circulating precursors compared with the administration of peptide alone, though not as great a decrease as that seen with IL-2. Immunization with peptide plus IL-2 did, however, appear to have clinical impact since 6 of the 16 patients (38%) that received peptide plus IL-2 had objective cancer regressions. It thus appeared possible that immunization with peptide plus IL-2 resulted in sequestering or apoptotic destruction of newly activated immune cells at the tumor site. These represent the first detailed studies of the impact of immunization with tumor peptides in conjunction with a variety of cytokines in patients with metastatic cancer.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Lipids , Melanoma/secondary , Melanoma/therapy , Adult , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Cell Line , Cytokines/administration & dosage , Cytokines/pharmacology , Freund's Adjuvant/administration & dosage , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Humans , Infusions, Intravenous , Injections, Subcutaneous , Interleukin-12/administration & dosage , Lymphocyte Activation , Melanoma/immunology , Membrane Glycoproteins/administration & dosage , Membrane Glycoproteins/immunology , Middle Aged , Neoplasm Proteins/administration & dosage , Neoplasm Proteins/immunology , Peptide Fragments/administration & dosage , Peptide Fragments/immunology , Peptides , Stem Cells/immunology , T-Lymphocytes/immunology , Tumor Cells, Cultured , gp100 Melanoma Antigen
13.
Blood ; 93(10): 3250-8, 1999 May 15.
Article En | MEDLINE | ID: mdl-10233876

This dose-escalation study was performed to evaluate the hematologic activity, biological effects, immunogenicity, and toxicity of PIXY321 (an interleukin-3/granulocyte-macrophage colony-stimulating factor fusion protein) administered after high-dose carboplatin (CBDCA) treatment. Patients with advanced cancers received CBDCA at 800 mg/m2 intravenously on day 0 of repeated 28-day cycles. In part A of the study, patients were treated with CBDCA alone during cycle 1 and then received PIXY321 on days 1 through 18 of cycle 2 and later cycles. In part B, patients received 18 days of PIXY321 beginning on day 1 of all CBDCA cycles, including cycle 1. PIXY321 was administered subcutaneously in 2 divided doses. Total doses of 135, 250, 500, 750, and 1,000 micrograms/m2/d were administered to successive cohorts of 3 to 6 patients in part A. In part B, patient groups received PIXY321 doses of 750, 1,000, and 1,250 micrograms/m2/d. The hematologic effects of PIXY321 were assessed in the first 2 cycles of therapy. Anti-PIXY321 antibody formation was assessed by enzyme-linked immunosorbent assay (ELISA) and neutralization assay. Of the 49 patients enrolled, 31 were fully evaluable for hematologic efficacy. When comparing the first B cycle (cycle B-1; with PIXY321) with the first A cycle (cycle A-1; without PIXY321), the fusion protein had no significant effect on platelet nadirs or duration of platelets less than 20,000/microL but was able to speed the time of recovery of platelet counts to 100,000/microL (15 v 20 days; P =.01). Significant improvements in neutrophil nadir and duration of ANC less than 500 were observed in cycles A-2 and B-1 (with PIXY321) as compared with cycle A-1 (without PIXY321). Initial PIXY321 prophylaxis (cycle A-2 and cycle B-1), enhanced the recovery of ANC to greater than 1,500/microL by an average of at least 8 days as compared with cycle A-1 (without PIXY321; P

Carboplatin/therapeutic use , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Interleukin-3/therapeutic use , Neoplasms/drug therapy , Adult , Antibody Formation , Carboplatin/adverse effects , Cholesterol/blood , Dose-Response Relationship, Drug , Drug Administration Schedule , Enzyme-Linked Immunosorbent Assay , Granulocyte-Macrophage Colony-Stimulating Factor/adverse effects , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacokinetics , Humans , Interleukin-3/adverse effects , Interleukin-3/immunology , Interleukin-3/pharmacokinetics , Leukocyte Count/drug effects , Platelet Count/drug effects , Recombinant Fusion Proteins/adverse effects , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/therapeutic use
14.
J Clin Oncol ; 16(8): 2752-60, 1998 Aug.
Article En | MEDLINE | ID: mdl-9704728

PURPOSE: We performed a phase I trial to determine whether in vivo expansion of activated CD4+ T cells was possible in cancer patients. 111Indium labeling was used to observe trafficking patterns of the infused stimulated CD4+ T cells. The influence of cyclophosphamide (CTX) dosing on immunologic outcome was also examined. PATIENTS AND METHODS: Patients with advanced solid tumors or non-Hodgkin's lymphoma received CTX at 300 or 1,000 mg/m2 intravenously (i.v.). Leukapheresis was performed to harvest peripheral-blood mononuclear cells (PBMCs) either just before the CTX dose, or when the patient was either entering or recovering from the leukocyte nadir induced by CTX. An enriched population of CD4+ T cells was obtained by negative selection. The CD4+ T cells were activated ex vivo with anti-CD3, cultured with interleukin-2 (IL-2) for 4 days, and adoptively transferred. After adoptive transfer, patients received IL-2 (9.0 x 10(6) IU/m2/d) by continuous infusion for 7 days. RESULTS: The absolute number of CD4+, CD4+/DR+, and CD4+/CD45RO+ T cells increased in a statistically significant fashion in all cohorts after the first course of therapy. The degree of CD4 expansion was much greater than CD8 expansion, which resulted in a CD4:CD8 ratio that increased in 26 of 31 patients. The greatest in vivo CD4 expansion occurred when cells were harvested as patients entered the CTX-induced nadir. One complete response (CR), two partial responses (PRs), and eight minor responses were observed. Trafficking of 111Indium-labeled CD4 cells to subcutaneous melanoma deposits was also documented. CONCLUSION: CD4+ T cells can be expanded in vivo in cancer patients, which results in increased CD4:CD8 ratios. The timing of pheresis in relation to CTX administration influences the degree of CD4 expansion. Tumor responses with this regimen were observed in a variety of tumors, including melanoma and non-Hodgkin's lymphoma; a high percentage of patients had at least some tumor regression from the regimen that produced the greatest CD4+ T-cell expansion.


Antineoplastic Agents/administration & dosage , CD3 Complex/immunology , CD4-Positive T-Lymphocytes/immunology , Cyclophosphamide/administration & dosage , Immunotherapy, Adoptive , Interleukin-2/administration & dosage , Lymphocyte Activation , Adult , Aged , Combined Modality Therapy , Female , Humans , Indium Radioisotopes , Infusions, Intravenous , Leukapheresis , Male , Middle Aged
16.
Semin Oncol ; 24(2): 173-86, 1997 Apr.
Article En | MEDLINE | ID: mdl-9129688

Differences in antigen expression between tumor cells and normal cells can be exploited to develop a variety of therapeutic anticancer agents. For example, vaccines containing tumor antigens can be administered to patients to elicit an immune-mediated attack against the tumor. Tumor antigens can also be targeted by passive transfer of monoclonal antibodies (MoAbs), or MoAbs that have been modified to carry toxin molecules. A large number of these antigen-specific agents are currently in clinical or late preclinical development. For the anticancer vaccines, much of the current development is focused on determining optimum immunization approaches. In the case of antibody-based reagents, second- and third-generation constructs are being developed to improve potency, decrease immunogenicity, and increase distribution to tumor.


Antineoplastic Agents , Cancer Vaccines , Drugs, Investigational , Immunotoxins , Animals , Antibodies, Bispecific , Antibodies, Monoclonal , Antigens, Neoplasm , Antineoplastic Agents/pharmacology , Cancer Vaccines/pharmacology , Humans , Immunotoxins/pharmacology , Neoplasms/drug therapy
18.
Clin Cancer Res ; 3(1): 39-46, 1997 Jan.
Article En | MEDLINE | ID: mdl-9815535

Interleukin 6 (IL-6) has antitumor activity comparable to IL-2 in murine models with less toxicity. Because the biological effects of intermittent and continuous infusions may differ, we conducted two concurrent Phase I trials of daily x5, 1-h, and continuous 120-h i.v. infusions to determine the toxicity, biological effects, and maximum tolerated dose of i.v. IL-6. Cohorts of six patients with advanced cancer received escalating doses (1, 3, 10, 30, 100, and 150 microgram/kg/day) of recombinant human IL-6 on days 1-5 and 8-12 of each 28-day course (1-h trial) or on days 1-5 of each 21-day course (120-h trial). Treatment was administered in regular inpatient wards and in outpatient clinics and was withheld in the event of grade 3 toxicity. Sixty-nine patients (1-h trial, n = 40; 120-h trial, n = 29) were enrolled, including 27 with renal cancer and 16 with melanoma. All were ambulatory, and 40 were asymptomatic. Fever (97%), anemia (78%), fatigue (56%), nausea or vomiting (49%), and elevated serum transaminase levels (42%) were the most frequent toxicities. Transient hypotension developed in 23 patients (33%). There were three deaths during the study due to progressive disease and/or infection. There were no objective responses. Dose-related increases in platelet counts and C-reactive protein levels were detected in most patients. Principal dose-limiting toxicities included atrial fibrillation (1 episode in the 1-h trial and 4 episodes in the 120-h trial) and neurological toxicities (3 episodes in the 1-h trial and 4 episodes in the 120-h trial). The neurological toxicities included confusion, slurred speech, blurred vision, proximal leg weakness, paraparesis, and ataxia. These effects were transient and reversed when IL-6 was discontinued. IL-6 can be given by i.v. infusion at biologically active doses with acceptable toxicity. Dose-limiting toxicities consisted mainly of a spectrum of severe but transient neurological toxicities and occasional episodes of atrial fibrillation. The maximum tolerated doses recommended for use with these i.v. schedules in Phase II trials are 100 microgram/kg/day by daily x5 1-h infusion and 30 microgram/kg/day by 120-h infusion. Phase II trials will be performed to determine the antitumor activity of IL-6 and better define its toxicity. Patients in these and other IL-6 studies should be monitored closely for neurological and cardiac effects.


Antineoplastic Agents/therapeutic use , Interleukin-6/therapeutic use , Neoplasms/drug therapy , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Cohort Studies , Dose-Response Relationship, Drug , Female , Humans , Injections, Intravenous , Interleukin-6/administration & dosage , Interleukin-6/adverse effects , Male , Middle Aged , Neurons/drug effects , Treatment Outcome
19.
Cancer J Sci Am ; 3 Suppl 1: S70-2, 1997 Dec.
Article En | MEDLINE | ID: mdl-9457398

PURPOSE: This article updates response duration and survival data for patients with metastatic renal cell carcinoma receiving high-dose recombinant interleukin-2 (rIL-2). PATIENTS AND METHODS: Two hundred fifty-five assessable renal cell carcinoma patients were entered into seven phase II clinical trials. They were administered 600,000 or 720,000 IU/kg rIL-2 by 15-minute intravenous infusion every 8 hours for up to 14 consecutive doses over 5 days as clinically tolerated with maximal support. A second identical cycle of treatment was scheduled following 5 to 9 days of rest, and courses could be repeated every 6 to 12 weeks in stable or responding patients. All data have been updated as of June 1996 with report forms completed by the clinical investigators. RESULTS: Objective overall responses have now been achieved in 37 of 255 patients (15%) with 17 complete (7%) and 20 partial (8%) responses. Median response duration for all objective responders is 54 months with a range of 3 to 107+ months. Median response duration for all complete responses has not been reached, with a range of 7 to 107+ months. Median response duration for all partial responses is 20 months, with a range of 3 to 97+ months. Median survival for all 255 patients was 16.3 months, and 10% to 20% of patients were estimated to be alive 5 to 10 years following treatment. CONCLUSION: Treatment with high-dose rIL-2 is extremely effective for a subset of patients with metastatic renal cell carcinoma.


Antineoplastic Agents/therapeutic use , Carcinoma, Renal Cell/therapy , Interleukin-2/analogs & derivatives , Kidney Neoplasms/therapy , Adolescent , Adult , Aged , Carcinoma, Renal Cell/mortality , Female , Humans , Interleukin-2/therapeutic use , Kidney Neoplasms/mortality , Male , Middle Aged , Recombinant Proteins/therapeutic use , Survival Rate , Treatment Outcome
20.
J Immunother Emphasis Tumor Immunol ; 19(5): 364-74, 1996 Sep.
Article En | MEDLINE | ID: mdl-8941876

We performed a prospective, randomized study to determine whether subcutaneous administration of interleukin-2 (IL-2) in combination with an autologous renal cell vaccine is feasible and can potentiate antitumor immunity. Seventeen patients with metastatic renal cell carcinoma underwent surgical resection with preparation of an autologous tumor cell vaccine. Patients were vaccinated intradermally twice at weakly intervals with 10(7) irradiated tumor cells plus bacillus Calmette-Guérin, and once with 10(7) tumor cells alone. Patients were randomized to one of three groups: no adjuvant IL-2, low-dose IL-2 (1.2 x 10(6) IU/m2), or high-dose IL-2 (1.2 x 10(7) IU/m2). IL-2 was administered subcutaneously on the day of vaccination and the subsequent 4 days. Immune response was monitored by delayed-type hypersensitivity (DTH) response to tumor cells as compared with normal autologous renal cells. Sixteen of 17 patients received vaccine therapy. Four patients developed cellular immunity specific for autologous tumor cells as measured by DTH responses; two had received no IL-2 and two had received high-dose IL-2. There were two partial responses (PR) noted, both in patients who received high-dose IL-2. One responding patient was DTH(+) and one was negative. A third patient who was DTH(+) after vaccination with no IL-2 had a dramatic PR after receiving IL-2 subcutaneously in a subsequent protocol. Prospective testing of response to recall antigens indicated that only 5 of 12 tested patients were positive, including both clinical responders. These data suggest that subcutaneously administered adjuvant IL-2 does not dramatically augment the immunologic response to autologous renal cell vaccines as determined by the development of tumor-specific DTH response.


Adjuvants, Immunologic/therapeutic use , Cancer Vaccines/therapeutic use , Carcinoma, Renal Cell/therapy , Interleukin-2/administration & dosage , Interleukin-2/therapeutic use , Kidney Neoplasms/therapy , Cancer Vaccines/administration & dosage , Cancer Vaccines/adverse effects , Carcinoma, Renal Cell/immunology , Chemotherapy, Adjuvant , Humans , Hypersensitivity, Delayed/immunology , Immunotherapy, Adoptive , Injections, Subcutaneous , Kidney Neoplasms/immunology
...