Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
1.
bioRxiv ; 2023 Apr 28.
Article in English | MEDLINE | ID: mdl-36896021

ABSTRACT

Hoefges et al. utilized a whole-proteome peptide array approach to show that C57BL/6 mice develop a large repertoire of antibodies against linear peptide sequences of their melanoma after receiving a curative immunotherapy regimen consisting of radiation and an immunocytokine. Antibodies can play an important role in innate and adaptive immune responses against cancer, and in preventing infectious disease. Flow cytometry analysis of sera of immune mice that were previously cured of their melanoma through a combined immunotherapy regimen with long-term memory showed strong antibody-binding against melanoma tumor cell lines. Using a high-density whole-proteome peptide array, we assessed potential protein-targets for antibodies found in immune sera. Sera from 6 of these cured mice were analyzed with this high-density, whole-proteome peptide array to determine specific antibody-binding sites and their linear peptide sequence. We identified thousands of peptides that were targeted by 2 or more of these 6 mice and exhibited strong antibody binding only by immune, not naive sera. Confirmatory studies were done to validate these results using 2 separate ELISA-based systems. To the best of our knowledge, this is the first study of the "immunome" of protein-based epitopes that are recognized by immune sera from mice cured of cancer via immunotherapy.

2.
Hematol Oncol Clin North Am ; 15(4): 703-21, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11676280

ABSTRACT

Preclinical and clinical development of antitumor strategies using mAbs are showing antitumor efficacy in animal models and in some clinical settings. Preclinical models suggest that mAb treatment would be most effective when provided in the minimal residual disease setting and can involve mAbs in a variety of roles. In murine models, the combination of mAbs with recombinant cytokines, such as IL-2, IL-12, or GM-CSF, can augment the immunologic effect of the mAbs by activating effector cell functions. Efficacy appears to be greatest when the mAb can recruit the effector cells of the host's immune system into helping in the mediation of the antitumor effect. Clinical testing of these concepts is under way.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Neoplasm/therapeutic use , Antibody-Dependent Cell Cytotoxicity , Neoplasms/therapy , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibodies, Neoplasm/immunology , Antigens, CD/immunology , Antigens, Neoplasm/immunology , Apoptosis , Bone Marrow Purging , Cancer Vaccines/therapeutic use , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Combined Modality Therapy , Complement Activation , Cytokines/therapeutic use , Drug Design , Genetic Engineering , Humans , Interleukin-2/genetics , Interleukin-2/therapeutic use , Lymphocyte Activation , Mice , Molecular Mimicry , Neoplasms/immunology , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/therapeutic use
3.
Cancer ; 88(8): 1892-901, 2000 Apr 15.
Article in English | MEDLINE | ID: mdl-10760767

ABSTRACT

BACKGROUND: Interleukin-2 (IL-2) and granulocyte-macrophage-colony stimulating factor (GM-CSF) are cytokines with nonoverlapping pleiotropic effects. In a prior Phase Ib study, this combination of agents exhibited antitumor effects in the lungs of four of eight patients with renal cell carcinoma and pulmonary metastases. We conducted this Phase Ib/II trial to determine the response rate of renal cell carcinoma patients with pulmonary metastases treated with continuous infusion IL-2 plus GM-CSF. METHODS: Patients with renal cell carcinoma and pulmonary metastases were treated with 1.5, 2.25, or 4.5 x 10(6) IU/m(2)/day 96-hour continuous infusion IL-2 on Days 1-4, 8-11, and 15-18, and 1.25, 2.25, or 2.5 microg/kg/day GM-CSF on Days 8-19. RESULTS: Sixteen patients were treated per protocol, 14 of whom could be evaluated for disease progression. None of these 14 patients had >50% shrinkage of either total tumor burden or pulmonary metastasis. One patient developed Grade 5 neurotoxicity. Autopsy revealed acute multifocal cerebral venous thrombosis as well as acute subdural and subarachnoid hemorrhage. CONCLUSIONS: The combination of IL-2 and GM-CSF may be associated with marked morbidity and, as in one case in this study, mortality. No significant antitumor activity was appreciated. Thus, the combination of IL-2 and GM-CSF, when administered at this dose and according to this schedule, does not appear to be active in renal cell carcinoma and is associated with significant toxicities. Further studies using this combination of agents should only be undertaken with extreme caution and particular attention to neurotoxicity.


Subject(s)
Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/secondary , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Interleukin-2/administration & dosage , Kidney Neoplasms/drug therapy , Kidney Neoplasms/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Adult , Aged , Cerebral Hemorrhage/chemically induced , Drug Administration Schedule , Drug Therapy, Combination , Fatal Outcome , Female , Granulocyte-Macrophage Colony-Stimulating Factor/adverse effects , Humans , Interleukin-2/adverse effects , Intracranial Thrombosis/chemically induced , Male , Middle Aged , Nervous System/drug effects , Treatment Outcome , Venous Thrombosis/chemically induced
4.
Cancer Immunol Immunother ; 48(5): 219-29, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10478638

ABSTRACT

The fusion protein formed from ch14.18 and interleukin-2 (ch14.18-IL-2), shown to exhibit antitumor efficacy in mouse models, consists of IL-2 genetically linked to each heavy chain of the ch14.18 chimeric anti-GD2 monoclonal antibody. The purpose of this study was to determine the pharmacokinetics of ch14.18-IL-2 in mice and assess its stability in murine serum. Following i.v. injection, the fusion protein was found to have a terminal half-life of 4.1 h. Detection of IL-2 following injection of the ch14.18-IL-2 fusion protein showed a similar half-life, indicating that the fusion protein prolongs the circulatory half-life of IL-2. Detection of human IgG1 following injection of ch14.18-IL-2 showed a terminal half-life of 26.9 h. These data suggested that the native fusion protein is being altered in vivo, resulting in a somewhat rapid loss of detectable IL-2, despite prolonged circulation of its immunoglobulin components. In vitro incubation of the ch14.18-IL-2 fusion protein in pooled mouse serum at 37 degrees C for 48 h resulted in a loss of its IL-2 component, as detected in enzyme-linked immunosorbent assay systems and in proliferation assays. Polyacrylamide gel electrophoresis and Western blot analysis of the fusion protein incubated in mouse serum at 37 degrees C indicated that the ch14.18-IL-2 is cleaved, resulting in a loss of the 67-kDa band (representing the IL-2 linked to the IgG1 heavy chain) and the detection of a band of more than 50 kDa, slightly heavier than the IgG1 heavy chain itself. This suggests that the fusion protein is being cleaved in vitro within the IL-2 portion of the molecule. These studies show that (1) ch14.18-IL-2 prolongs the circulatory half-life of IL-2 (compared to that of soluble IL-2) and (2) the in vivo clearance of the fusion protein occurs more rapidly than the clearance of the ch14.18 antibody itself, possibly reflecting in vivo cleavage within the IL-2 portion of the molecule, resulting in loss of IL-2 activity.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacology , Interleukin-2/chemistry , Interleukin-2/pharmacokinetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacokinetics , Animals , Antibodies, Monoclonal/blood , Antibodies, Monoclonal/pharmacokinetics , Drug Stability , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Humans , Immunization, Passive/methods , Interleukin-2/blood , Mice , Mice, Inbred BALB C , Recombinant Fusion Proteins/blood , Temperature , Time Factors
5.
Clin Diagn Lab Immunol ; 6(2): 236-42, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10066660

ABSTRACT

Preliminary testing has shown in vitro and in vivo that antitumor activity can be obtained with fusion proteins linking tumor-reactive monoclonal antibodies to cytokines, such as granulocyte-macrophage colony-stimulating factor or interleukin 2 (IL-2). Preclinical and clinical testing of these reagents requires their in vitro and in vivo quantitation and pharmacokinetic evaluation. We have focused on the detection of a fusion protein which links one human IL-2 molecule to the carboxy terminus of each heavy chain of the tumor-reactive human-mouse chimeric anti-GD2 antibody, ch14.18. We have developed enzyme-linked immunosorbent assays (ELISAs) to evaluate intact tumor-reactive fusion proteins. By these ELISAs we can reliably measure nanogram quantities of intact ch14.18-IL-2 fusion protein and distinguish the intact protein from its components (ch14.18 and IL-2) in buffer, mouse serum, and human serum with specificity and reproducibility. The measurement of intact ch14.18-IL-2 fusion protein is not confounded by free IL-2 or free ch14.18 when 100 ng or less of total immunoglobulin per ml is used during the assay procedure. Our results indicate that these ELISAs are suitable for preclinical and clinical testing and with slight modifications are applicable to the analysis of a variety of other fusion proteins.


Subject(s)
Enzyme-Linked Immunosorbent Assay/methods , Granulocyte-Macrophage Colony-Stimulating Factor/analysis , Immunoglobulin G/analysis , Interleukin-2/analysis , Recombinant Fusion Proteins/analysis , Animals , Antibodies, Monoclonal , Binding, Competitive/immunology , Blood Proteins , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay/standards , Epitopes , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Humans , Immunoglobulin G/immunology , Indicator Dilution Techniques , Interleukin-2/immunology , Mice , Recombinant Fusion Proteins/immunology , Reproducibility of Results
6.
Clin Cancer Res ; 5(2): 281-9, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10037176

ABSTRACT

Interleukin-2 (IL-2) is a potent lymphokine that activates natural killer cells, T cells, and other cells of the immune system. Several distinct recombinant human IL-2 preparations have shown antitumor activity, particularly for renal cell cancer and melanoma. Somewhat distinct immune and clinical effects have been noted when different IL-2 preparations have been tested clinically; however, the regimens and doses used were not identical. To compare these more directly, we have evaluated two clinical recombinant IL-2 preparations in vitro and in vivo using similar regimens and similar IUs of IL-2. We used the Food and Drug Administration-approved, commercially available Chiron IL-2 and the Hoffmann LaRoche (HLR) IL-2 supplied by the National Cancer Institute. Using equivalent IUs of IL-2, we noted quantitative differences in vitro and in vivo in the IL-2 activity of these two preparations. In patients receiving comparable IUs of the two preparations, HLR IL-2 induced the release of more soluble IL-2 receptor alpha into the serum than Chiron IL-2. In addition, more toxicities were noted in patients receiving 1.5 x 10(6) IU of HLR IL-2 than were seen in patients treated with 1.5 x 10(6) or even 4.5 x 10(6) IU of Chiron IL-2. These toxicities included fever, nausea and vomiting, and hepatic toxicity. In vitro proliferative assays using IL-2-dependent human and murine cell lines indicated that the IU of HLR IL-2 was more effective than Chiron IL-2 at inducing tritiated thymidine incorporation. Using flow cytometry, we also found quantitative differences in the ability of these two preparations to bind to IL-2 receptors. These findings indicate that approximately 3-6 IU of Chiron IL-2 are required to induce the same biological effect as 1 IU of HLR IL-2.


Subject(s)
Interleukin-2/therapeutic use , Neoplasms/drug therapy , Animals , CD56 Antigen/immunology , Cell Division/drug effects , Humans , Infusion Pumps , Interleukin-2/adverse effects , Interleukin-2/pharmacology , Killer Cells, Natural/immunology , Lymphocytosis/chemically induced , Mice , Neoplasms/immunology , Receptors, Interleukin-2/metabolism , Recombinant Proteins/adverse effects , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Reference Standards , Serum Albumin/pharmacology , Tumor Cells, Cultured
8.
Cancer Immunol Immunother ; 46(6): 327-37, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9756417

ABSTRACT

Administration of the cytokine interleukin-2 (IL-2) can result in therapeutic benefits for individuals with renal cell carcinoma and melanoma. Here we report an analysis of the transcription factor families AP-1, Sp1, NF-kappaB, and signal transducers and activators of transcription (STAT) in cancer patients' lymphocytes before and after IL-2 immunotherapy, as assessed by a gel-shift assay. An in vitro surrogate of IL-2 immunotherapy is the incubation of fresh peripheral blood mononuclear cells (PBMC) from healthy individuals in IL-2 for several days, resulting in the production of lymphokine-activated killer (LAK) activity in these cultures. One purpose of this study was to describe the profile of transcription factor activation in these different populations, and assess whether the patterns observed correlated with functional differences in these cells. Prior to in vivo IL-2 administration, the typical binding pattern of transcription factors in PBMC from patients resembled that seen in fresh PBMC from healthy individuals. Over a 3-week course of IL-2 therapy, in most patients the binding patterns of AP-1 , Sp1, and NF-kappaB proteins changed to resemble those seen in PBMC activated by IL-2 in vitro. However, the cells obtained from IL-2-treated patients did not have low-level constitutive expression of STAT binding factors as did LAK cells. When these patient cells were further stimulated by IL-2 in vitro, additional differences in STAT induction patterns were noted. These data provide further information on the molecular events occurring in immune cells generated through in vivo and in vitro administration of IL-2, and further document that there is not a precise congruence between PBMC activated in vivo and in vitro by IL-2.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , Immunologic Factors/pharmacology , Immunotherapy , Interleukin-2/pharmacology , Killer Cells, Lymphokine-Activated/drug effects , Lymphocytes/drug effects , Melanoma/therapy , Signal Transduction/drug effects , Transcription Factors/metabolism , Antibodies, Monoclonal/therapeutic use , Antibodies, Neoplasm/therapeutic use , Cells, Cultured , DNA-Binding Proteins/metabolism , Humans , Immunologic Factors/therapeutic use , Interleukin-2/therapeutic use , Killer Cells, Lymphokine-Activated/metabolism , Lymphocytes/metabolism , Melanoma/immunology , NF-kappa B/metabolism , STAT1 Transcription Factor , STAT2 Transcription Factor , Sp1 Transcription Factor/metabolism , Trans-Activators/metabolism , Transcription Factor AP-1/metabolism , Transcription Factors/classification
9.
Clin Cancer Res ; 4(5): 1287-96, 1998 May.
Article in English | MEDLINE | ID: mdl-9607589

ABSTRACT

Lymphocytes from patients receiving in vivo interleukin (IL)-2 therapy possess enhanced in vitro proliferative and cytotoxic responses to IL-2. The cells from these patients that respond to exogenous IL-2 are CD56+ natural killer cells expressing intermediate-affinity IL-2 receptor betagamma(c) complexes. Because IL-15 activates cells via these same betagamma(c) receptors, we hypothesized that IL-15 would also activate lymphocytes from patients treated with in vivo IL-2 therapy and therefore that IL-15 might potentially be useful as an immunotherapeutic agent alone or in combination with IL-2. We report here that peripheral blood mononuclear cells (PBMCs) from patients receiving in vivo IL-2 therapy do proliferate in response to IL-15. However, a greater dose of IL-15 is needed to reach the same level of proliferation stimulated by IL-2. The EC50 for IL-2 is 0.21 +/- 0.04 nM (mean +/- SE; n = 18), whereas the EC50 for IL-15-stimulated proliferation is 1.16 +/- 0.16 nM (n = 18). In contrast to the proliferative response, equivalent doses of IL-2 and IL-15 stimulate patient PBMCs to mediate similar levels of cytotoxicity against Daudi, K562, and LA-N-5 tumor targets. Notably, low concentrations of IL-15 that do not stimulate a substantial proliferative response (e.g., 1.0 ng/ml) do boost PBMCs to mediate cytotoxicity against these tumor targets. These distinct dose-response curves for proliferation compared to cytotoxicity suggest that IL-15 should be evaluated for its potential as an immunotherapeutic agent to treat cancer, particularly in regimens providing doses that might minimize the proliferative response (associated with cytokine release and toxic side effects) while maintaining the cytolytic antitumor response.


Subject(s)
Interleukin-15/pharmacology , Interleukin-2/pharmacology , Lymphocyte Subsets/drug effects , Melanoma/immunology , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , CD56 Antigen/metabolism , Cell Division/drug effects , Dose-Response Relationship, Drug , Humans , Interleukin-2/therapeutic use , Killer Cells, Natural/cytology , Killer Cells, Natural/drug effects , Lectins, C-Type , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Lymphocyte Subsets/cytology , Melanoma/drug therapy , Tumor Cells, Cultured
10.
Clin Cancer Res ; 3(8): 1277-88, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9815810

ABSTRACT

We conducted a Phase IB trial of antidisialoganglioside chimeric 14. 18 (ch14.18) antibody and interleukin 2 (IL-2) to determine the maximal tolerated dose (MTD), immunological effects, antitumor effects, and toxicity of this treatment combination. Twenty-four melanoma patients received immunotherapy with ch14.18 antibody and a continuous infusion of Roche IL-2 (1.5 x 10(6) units/m2/day) given 4 days/week for 3 weeks. The ch14.18 antibody (dose level, 2-10 mg/m2/day) was scheduled to be given for 5 days, before, during, or following initial systemic IL-2 treatment. The ch14.18 MTD was 7.5 mg/m2/day, and 15 patients were treated with the ch14.18 MTD. Immunological effects included the induction of lymphokine-activated killer activity and antibody-dependent cellular cytotoxicity by peripheral blood mononuclear cells. In addition, serum samples obtained following ch14.18 infusions were able to facilitate in vitro antibody-dependent cellular cytotoxicity. Antitumor activity included one complete response, one partial response, eight patients with stable disease, and one patient with >50% decrease of hepatic metastases in the face of recurrence of a s.c. lesion. Dose-limiting toxicities were a severe allergic reaction and weakness, pericardial effusion, and decreased performance status. Most patients treated at the MTD had abdominal, chest, or extremity pain requiring i.v. morphine. One patient had an objective peripheral neuropathy. This IL-2 and ch14.18 treatment combination induces immune activation in all patients and antitumor activity in some melanoma patients. We are attempting to enhance this treatment approach by addition of the anti-GD3 R24 antibody to this IL-2 and ch14.18 regimen.


Subject(s)
Antibodies, Monoclonal/adverse effects , Interleukin-2/adverse effects , Melanoma/therapy , Adult , Antibodies, Anti-Idiotypic/blood , Antibody-Dependent Cell Cytotoxicity , Dose-Response Relationship, Drug , Female , Humans , Immunophenotyping , Immunotherapy , Killer Cells, Lymphokine-Activated/immunology , Lymphocyte Count , Lymphocytes/immunology , Male , Melanoma/immunology , Melanoma/pathology , Middle Aged , Neoplasm Metastasis , Recombinant Fusion Proteins/adverse effects , Recombinant Proteins/adverse effects , Tumor Cells, Cultured
11.
Cancer ; 80(2): 317-33, 1997 Jul 15.
Article in English | MEDLINE | ID: mdl-9217046

ABSTRACT

BACKGROUND: The murine monoclonal antibody (MoAb) 14.G2a recognizes GD2, a disialoganglioside expressed in tumors of neuroectodermal origin, and facilitates antibody dependent cellular cytotoxicity (ADCC) in vitro. When given in vivo, interleukin-2 (IL-2) can increase ADCC by enhancing the activity and number of circulating lymphocytes. METHODS: Thirty-three pediatric patients with GD2 positive malignancies, ranging in age from 2 to 17 years (median, 9.9 years), received IL-2 and 14.G2a in this Phase I/IB study of the Children's Cancer Group (CCG) and were monitored for toxicities and response to therapy. Seven of these patients also received granulocyte-macrophage-colony stimulating factor. RESULTS: The maximum tolerated dose (MTD) of 14.G2a with IL-2 was 15 mg/m2/day. The most prevalent Grade 3-4 toxicities were generalized pain (n = 14 [42%]) and fever without documented infection (n = 17 [52%]). IL-2 was thought to be the causative agent in most cases of fever. Toxicities attributed to 14.G2a included pain, allergic or anaphylactic reactions, and rash. Human antimouse antibodies were demonstrated in 9 of 21 evaluated patients. One patient with neuroblastoma had a partial response, and one patient with osteosarcoma had a complete response. Immunocytology demonstrated that the number of neuroblastoma cells in bone marrow decreased in three patients. CONCLUSIONS: The murine MoAb 14.G2a was well tolerated at the MTD and appeared to have some antitumor activity. Further development of this approach will involve additional engineered forms of the antibody as well as testing in the adjuvant and minimal residual disease setting.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Antibodies, Monoclonal/therapeutic use , Gangliosides/immunology , Interleukin-2/therapeutic use , Neoplasm Proteins/immunology , Neuroblastoma/therapy , Adjuvants, Immunologic/adverse effects , Adolescent , Animals , Antibodies, Monoclonal/adverse effects , Antibody-Dependent Cell Cytotoxicity , Bone Marrow/pathology , Child , Child, Preschool , Female , Humans , Interleukin-2/adverse effects , Male , Mice , Neuroblastoma/pathology , Recombinant Proteins/therapeutic use , Remission Induction
12.
Cancer J Sci Am ; 3 Suppl 1: S121-7, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9457407

ABSTRACT

PURPOSE: Administration of recombinant interleukin-2 (rIL-2) causes activation of natural killer (NK) cells, which express CD16, the Fc receptor gamma III (Fc gamma III), and can mediate antibody-dependent cellular cytotoxicity (ADCC). In an effort to generate a more directed antitumor response by rIL-2-activated NK cells, we have investigated combination therapy with rIL-2 plus the antitumor monoclonal antibody (mAb) 14. G2A and its molecular derivatives, which react strongly with the GD2 ganglioside expressed on melanoma, neuroblastoma, and certain other tumors. PATIENTS AND METHODS: The initial trial of this therapeutic strategy involved 33 evaluable neuroblastoma patients who received rIL-2 by continuous intravenous infusion in combination with the murine 14.G2A mAb. A follow-up phase I study was conducted in 24 evaluable adult melanoma patients with a mouse/human chimeric mAb (ch14.18). The ch14.18 mAb has subsequently been investigated in combination with rIL-2 and granulocyte-macrophage colony-stimulating factor in neuroblastoma patients following autologous bone marrow transplantation. Based on preclinical data, the administration of rIL-2 plus mAbs may be more effective in patients with minimal residual disease. An alternative strategy to induce ADCC has also been investigated because of the finding that many NK cells fail to express Fc gamma III. The IL-2-ch14.18 fusion protein has been tested in preclinical studies for its ability to induce an effective antitumor response and is currently being evaluated for future clinical testing. RESULTS: Based on in vitro assays, the serum of patients treated with rIL-2 plus either 14.G2A or ch14.18 mAbs demonstrated sufficient levels of antibody and NK cell activity to mediate effective ADCC; however, many patients developed antibodies against the administered murine and chimeric mAbs, leading to allergic complications. Concurrent administration of rIL-2 and ch14.18 resulted in a lower incidence of anti-ch14.18 antibodies. The IL-2-ch14.18 fusion protein has demonstrated dramatic antitumor effects in murine models. DISCUSSION: These studies have demonstrated the feasibility of concurrent administration of rIL-2 with the mouse/human chimeric ch14.18 mAb. Although the early pilot studies were conducted in patients with bulky disseminated disease, this approach may be more effective in patients with minimal residual disease. Further refinement of the reagents and phase II and III trials will be necessary to evaluate fully the safety and efficacy of this immunotherapeutic approach.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Immunotherapy , Interleukin-2/administration & dosage , Neuroblastoma/therapy , Cytokines/administration & dosage , Dose-Response Relationship, Immunologic , Humans , Lymphocyte Count , Treatment Outcome
13.
Clin Cancer Res ; 2(12): 1951-9, 1996 Dec.
Article in English | MEDLINE | ID: mdl-9816154

ABSTRACT

Cytotoxic effector cells interact with target cells through various mechanisms. CTLs use the antigen-specific T cell receptor, whereas Fc receptor-positive natural killer cells use this receptor to interact with antibody-coated target cells. We evaluated the tumor-binding and lymphocyte-activating capability of a recombinant fusion protein consisting of a tumor-selective human/mouse chimeric anti-ganglioside GD2 antibody (ch14.18) and recombinant human interleukin-2 (IL2) (ch14.18-IL2). This fusion protein bound specifically to GD2-positive melanoma and neuroblastoma tumor cell lines, and its IL2 component stimulated in vitro proliferation of an IL2-dependent cell line, as well as peripheral blood mononuclear cells, in healthy control individuals and in cancer patients receiving continuous infusion of IL2. The IL2 presented by the fusion protein, when bound to tumor cells, induced proliferation of IL2-responsive cells as well as a comparable amount of soluble IL2 did. This suggests that localization of IL2 at the site of contact between tumor and effector cells is an effective way of presenting this cytokine to IL2-responsive cells. The ch14.18-IL2 fusion protein also mediated antibody-dependent cellular cytotoxicity with Fc receptor-positive effector cells to an extent similar to ch14.18. These results, together with those of previous studies documenting antitumor efficacy against human tumor xenografts in SCID mice and GD2-positive murine tumors in immunocompetent syngeneic mice, suggest that the ch14.18-IL2 fusion protein should be tested in Phase I and II trials in patients with GD2-positive tumors.


Subject(s)
Gangliosides/immunology , Immunotoxins/therapeutic use , Interleukin-2/pharmacology , Killer Cells, Natural/drug effects , Lymphocyte Activation/drug effects , Animals , Antibodies/genetics , Antibodies/therapeutic use , Antibody-Dependent Cell Cytotoxicity/immunology , Cell Division/drug effects , Dose-Response Relationship, Drug , Flow Cytometry , Humans , Immunotoxins/genetics , Interleukin-2/metabolism , Killer Cells, Natural/immunology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Lymphocyte Activation/immunology , Mice , Receptors, Interleukin-2/immunology , Receptors, Interleukin-2/metabolism , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Tumor Cells, Cultured
14.
J Immunother Emphasis Tumor Immunol ; 19(4): 278-95, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8877722

ABSTRACT

The induction of human antimouse antibodies (HAMA) and human anti-idiotypic (anti-Id) responses in cancer patients receiving therapeutic monoclonal antibody (mAb) may limit the effectiveness of the administered mAb. This report evaluates the influence of systemic interleukin-2 (IL-2) on the anti-Id response to anti-disialoganglioside (anti-GD2) antibody given as treatment for patients with melanoma. Twenty-eight patients with melanoma received combined immunotherapy with anti-GD2 antibody and IL-2 at 1.5 x 10(6) U/m2/day given 4 days/week. The anti-GD2 antibody [murine 14.G2a mAb; dose levels of 2-5 mg/m2/day (4 patients); or human-mouse chimeric 14.18 (ch14.18) antibody; dose levels of 2-10 mg/m2/day (24 patients)] was scheduled to be given for 5 days either before, during, or after initial systemic IL-2 treatment. All four patients who received murine 14.G2a developed HAMA anti-isotype antibodies (660-1,000 ng/ml) as well as measurable anti-Id antibodies. All three patients who received initial treatment with ch14.18 alone developed a strong anti-Id antibody response after IL-2 was started 1 week later. The serum level of anti-Id antibody decreased during subsequent ch14.18 infusions, suggesting that the anti-Id antibody may be binding the administered ch14.18. In contrast, measurable anti-Id antibody was detected in only 3 of 14 patients who received IL-2 before, during, and after initial ch14.18 administration. Two of four patients receiving systemic IL-2 before and during initial ch14.18 infusions, and two of three patients receiving systemic IL-2 concurrent with initial ch14.18 infusions developed anti-Id antibodies. These data suggest that the anti-Id response to chimeric anti-GD2 antibody is influenced by the timing of systemic IL-2 in relation to antibody administration and can be suppressed by systemic treatment with IL-2 given before, during, and after the antibody administration.


Subject(s)
Adjuvants, Immunologic/pharmacology , Antibodies, Anti-Idiotypic/biosynthesis , Antibodies, Monoclonal/therapeutic use , Gangliosides/immunology , Immunoglobulin Idiotypes/immunology , Interleukin-2/pharmacology , Melanoma/immunology , Antibodies, Anti-Idiotypic/drug effects , Antibodies, Monoclonal/immunology , Antibody-Dependent Cell Cytotoxicity , Drug Synergism , Humans , Infusions, Intravenous , Interleukin-2/administration & dosage , Interleukin-2/therapeutic use , Melanoma/therapy , Recombinant Proteins/administration & dosage , Recombinant Proteins/immunology , Recombinant Proteins/therapeutic use , Skin Neoplasms/immunology , Skin Neoplasms/therapy
15.
J Immunother Emphasis Tumor Immunol ; 19(3): 184-91, 1996 May.
Article in English | MEDLINE | ID: mdl-8811493

ABSTRACT

Renal cell carcinoma (RCC) is relatively resistant to chemotherapy and radiotherapy, whereas treatment with biologics has achieved limited success. Although monoclonal antibodies able to recognize human RCC have been identified, most induce little complement-dependent cytotoxicity or antibody-dependent cellular cytotoxicity (ADCC), and thus are of limited potential as therapeutic modalities in their natural conformation. We evaluated a human/ mouse chimeric derivative of the previously described G250 murine monoclonal antibody (mAb), reactive with RCC, to identify a reagent for potential immunotherapy. This chimeric antibody (ch-G250) is composed of the murine variable region from the G250 mAb, which recognizes a tumor-associated antigen expressed on 95% of primary and 86% of metastatic renal cell carcinomas. The constant region of the ch-G250 is comprised of the human IgG1 isotype domains. This chimeric antibody does not bind to normal renal tissue or other normal human tissues, with the exception of gastric mucosal cells and large bile-duct epithelium. Clinical radiolocalization studies have demonstrated the relative tumor-targeting potential of this radiolabeled antibody. This ch-G250 antibody facilitated potent ADCC against several RCC lines when using in vitro and in vivo interleukin-2 (IL-2)-activated peripheral blood mononuclear cells obtained from healthy control donors and patients with cancer, respectively. This lymphocyte-mediated ADCC was specific for RCC cells recognized by the ch-G250 antibody. Using flow cytometry, we found that the level of ADCC was directly related to the degree of binding of ch-G250 to the renal cell target. These in vitro data suggest that this antibody may improve efficacy of IL-2 therapy by targeting cytokine-activated effector cells directly to the tumor and facilitating in vivo ADCC. Clinical studies combining this chimeric antibody with IL-2 treatment will be needed to test the antitumor effects of this ADCC effect in vivo.


Subject(s)
Antibodies, Neoplasm/therapeutic use , Antibody-Dependent Cell Cytotoxicity/drug effects , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/therapy , Interleukin-2/therapeutic use , Kidney Neoplasms/therapy , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/therapeutic use , Animals , Cells, Cultured , Flow Cytometry , Humans , Immunization, Passive/methods , Interleukin-2/immunology , Kidney Neoplasms/immunology , Mice
16.
Clin Cancer Res ; 2(2): 319-30, 1996 Feb.
Article in English | MEDLINE | ID: mdl-9816175

ABSTRACT

Interleukin 2 (IL-2) and granulocytes-macrophage colony-stimulating factor (GM-CSF) are activators of the lymphocyte and granulocyte/macrophage series, respectively. We conducted a phase IB trial to identify the maximally tolerated dose and to assess immunological effects of the combination. Thirty-four patients with incurable cancers received 2.5, 5, or 10 microgram/kg GM-CSF s.c. either before or concurrently with 1.5 or 3.0 million units/m2/day IL-2. The most common laboratory and clinical side effects included an elevation of the total WBC or eosinophil count due to GM-CSF, and constitutional symptoms due to IL-2. Grade 3 or 4 toxicities included hypotension, thrombocytopenia, elevations in aspartate aminotransferase or bilirubin, renal toxicity, gastrointestinal hemorrhage, arrhythmia, and constitutional symptoms. Two patients receiving 5.0 microgram/kg GM-CSF plus concurrent 3.0 million units IL-2 experienced dose-limiting grade 3 or 4 neurological toxicity, which reversed almost completely. An increase in the serum-soluble IL-2 alpha chain receptor was observed with administration of GM-CSF, IL-2, or the combination. IL-2 therapy enhanced lymphokine-activated killer activity, antibody-dependent cellular cytotoxicity, and lymphocyte activation, with increased CD16 and CD56 expression. GM-CSF increased expression of human leukocyte antigen DR on peripheral blood monocytes and decreased surface expression of CD16 on circulating monocytes and polymorphonuclear cells. Lymphokine-activated killer activity and CD16 expression on monocytes and lymphocytes and CD56 expression on lymphocytes were significantly lower in patients receiving GM-CSF simultaneously with IL-2 than in patients receiving the sequential treatment. Antitumor activity was observed in the lungs of four of eight renal cell carcinoma patients with pulmonary metastases treated with concurrent GM-CSF and IL-2. Although no or minimal shrinkage was observed in the patients' large primary tumors, these results warrant further study. The recommended initial Phase II dose and schedule is 1.25 microgram/kg/day GM-CSF, given concurrently with 1.5 million Roche units/m2/day (4.5 x 10(6) international units/m2/day) IL-2, with subsequent escalation of GM-CSF to 2.5 microgram/kg/day after careful observation for toxicities.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Interleukin-2/administration & dosage , Neoplasms/therapy , Adult , Aged , Antibody-Dependent Cell Cytotoxicity , CD56 Antigen/analysis , Female , Granulocyte-Macrophage Colony-Stimulating Factor/adverse effects , HLA-DR Antigens/analysis , Humans , Interleukin-2/adverse effects , Killer Cells, Lymphokine-Activated/immunology , Male , Middle Aged , Neoplasms/immunology , Receptors, IgG/analysis , Receptors, Interleukin-2/analysis
17.
Cell Immunol ; 167(1): 99-107, 1996 Jan 10.
Article in English | MEDLINE | ID: mdl-8548851

ABSTRACT

Because certain antiganglioside monoclonal antibodies can facilitate antibody-dependent cellular cytotoxicity against GD2+ ganglioside-bearing human and canine tumor cells, we wished to determine if clinically relevant antiganglioside monoclonal antibodies (Mabs) could also fix canine complement to lyse tumor cells in vitro. Using flow cytometry, human tumor cell lines (M21 melanoma and OHS osteosarcoma) were shown to highly express ganglioside GD2 and, to a lesser degree, GD3. In 51Cr release assays, M21 cells were lysed with canine serum, as a source of complement, plus either Mab 14.G2a or its mouse-human chimera, ch 14.18, specific for GD2. Heating canine serum abrogated its lytic activity and addition of rabbit complement reconstituted M21 lysis. Similar results were obtained with M21 cells when Mab R24 (against GD3) and canine serum were used. OHS cells were also lysed with canine serum plus Mab 14.G2a and lytic activity was abolished by heating canine serum but reconstituted with rabbit complement. Alone, canine serum or Mabs were not lytic to M21 or OHS cells. Conversely, human neuroblastoma (LAN-5) and K562 erythroleukemia cells were lysed by canine serum alone which was shown by flow cytometry to contain naturally occurring canine IgM antibodies that bound LAN-5 and K562 cells. The lytic activity of canine serum for LAN-5 or K562 cells was abolished by heating and restored by addition of either human or rabbit complement. Thus, human tumor cell lines can be lysed with antiganglioside Mabs through fixation and activation of canine complement-dependent lytic pathways. Canine xenoantibodies also mediate complement-dependent cytotoxicity of some human tumor cell lines. Together, these results are significant because they demonstrate an antitumor effect of the canine immune system which is of potential importance for cancer immunotherapy in a promising animal model.


Subject(s)
Antibodies, Heterophile/immunology , Antibodies, Monoclonal/immunology , Complement System Proteins/physiology , Cytotoxicity, Immunologic , Gangliosides/physiology , Neoplasms/therapy , Animals , Antibodies, Monoclonal/therapeutic use , Dogs , Female , Flow Cytometry , Humans , Immunoglobulin G/classification , Immunoglobulin G/immunology , Male , Mice , Neoplasms/immunology , Rabbits , Tumor Cells, Cultured
18.
Blood ; 86(2): 636-45, 1995 Jul 15.
Article in English | MEDLINE | ID: mdl-7605993

ABSTRACT

We identified a dog with large granular lymphocytic leukemia and cutaneous lymphoma that exhibited constitutive expression of interleukin-2 (IL-2) receptors by the leukemic peripheral blood lymphocytes. The leukemic cells phenotypically resembled natural killer (NK) cells, and their surface IL-2 receptors were functional, as determined by the capacity to bind human recombinant IL-2 with high-affinity resulting in the transduction of proliferation signals and in the development of lymphokine-activated killer cell activity. These cells produced IL-2 spontaneously, and they may have maintained their proliferative state through an IL-2-dependent autocrine growth pathway. Our results indicate that neoplastic lymphocytes of syndromes that involve circulating leukemic cells with dermotropism can originate from NK-like cells. Additionally, the data also suggest that proliferative conditions such as these may be the result of the aberrant production of IL-2. Further, this case illustrates the potential for the use of hematopoietic malignancies in the dog as a suitable animal model for immune targeting of IL-2 receptors as a novel treatment approach for similar malignancies of human beings.


Subject(s)
Dog Diseases/immunology , Killer Cells, Natural/pathology , Lymphoma, Large B-Cell, Diffuse/veterinary , Lymphoproliferative Disorders/veterinary , Neoplasm Proteins/analysis , Neoplastic Stem Cells/chemistry , Receptors, Interleukin-2/analysis , Skin Neoplasms/veterinary , Animals , Cytotoxicity, Immunologic , Dogs , Female , Humans , Immunophenotyping , Interleukin-2/metabolism , Killer Cells, Natural/immunology , Lymphoma, Large B-Cell, Diffuse/immunology , Lymphoma, Large B-Cell, Diffuse/pathology , Lymphoproliferative Disorders/immunology , Lymphoproliferative Disorders/pathology , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/pathology , Receptors, Interleukin-2/metabolism , Recombinant Proteins/metabolism , Skin Neoplasms/immunology , Skin Neoplasms/pathology
19.
Cancer ; 75(12): 2959-65, 1995 Jun 15.
Article in English | MEDLINE | ID: mdl-7773948

ABSTRACT

BACKGROUND: Recombinant human Interleukin-2 (IL-2) has been effective at inducing measurable antitumor responses in adults with renal cell carcinoma and melanoma. It also is being tested as adjuvant therapy for patients with acute myeloid leukemia after autologous bone marrow transplantation. METHODS: The authors tested IL-2 in a pediatric Phase II trial using a regimen that has antitumor effects in adults and was proven to be tolerated acceptably in a prior Phase I pediatric trial. Thirty-eight patients were entered into this study of whom 36 received IL-2 and were evaluable (20 with sarcoma, 9 with neuroblastoma, 5 with renal cell carcinoma, and 1 each with melanoma and lymphoma). RESULTS: Interleukin-2 dose modifications were based on tolerance and toxicity, such that 46% of these patients received a 50% increase in IL-2 dose during the second week, and 81% of those receiving the elevated dose continued receiving this dose level during the third week of treatment. A single patient with renal cell carcinoma had a complete response that was maintained; the remaining 35 patients did not show objective evidence of tumor response sufficient to qualify as either a complete response or a partial response. CONCLUSIONS: Absolute lymphocyte counts were indicative of the immunostimulatory effect of this IL-2 regimen as observed for adults, with a median 7.2-fold increase. Nevertheless, despite immune activation, a sufficient number of patients were evaluated, indicating that IL-2 does not have measurable antitumor effects in children with large refractory sarcomas or neuroblastomas, whereas one of five children with renal cell carcinoma had a complete response, consistent with the 10-20% response rate observed in adults.


Subject(s)
Hodgkin Disease/therapy , Interleukin-2/administration & dosage , Kidney Neoplasms/therapy , Melanoma/therapy , Neuroblastoma/therapy , Sarcoma/therapy , Adolescent , Carcinoma, Renal Cell/therapy , Child , Drug Administration Schedule , Female , Humans , Infusions, Intravenous , Interleukin-2/toxicity , Lymphocyte Count , Male , Recombinant Proteins/administration & dosage , Recombinant Proteins/toxicity
20.
Clin Cancer Res ; 1(5): 481-91, 1995 May.
Article in English | MEDLINE | ID: mdl-9816007

ABSTRACT

Anti-CD3 mAb and interleukin 2 (IL-2) were used in a Phase I study to treat 29 patients with cancer. The anti-CD3 was given as an i.v. bolus infusion over 10 min followed by two i.v. 96-h continuous infusions of IL-2 at 3 x 10(6) units/m2/day with a 3-day rest between the IL-2 infusions. Four patients were treated with 6, 18, 60, and 300 microgram/m2 anti-CD3. One patient received 3000 microgram/m2 anti-CD3. This patient developed profound hypotension and the IL-2 infusions were delayed for 2 weeks. Two patients were treated at an intermediate dose of 600 microgram/m2. These patients developed dose-limiting toxicities including hypotension, dyspnea and increased blood urea nitrogen, creatinine, and bilirubin. They were unable to complete their first course of therapy. In an effort to achieve a dose of anti-CD3 which would activate T cells in vivo, pentoxifylline was given to blunt the toxicities seen with anti-CD3 thought to be due predominantly to the cytokine syndrome and tumor necrosis factor release. Four patients received p.o. pentoxifylline to cover an anti-CD3 dose of 600 microgram/m2. The IL-2 infusion was initiated 1 week after the mAb. While there was an anti-CD3 dose-dependent increase in serum tumor necrosis factor level 1 h after mAb infusion, pentoxifylline did not reduce the serum tumor necrosis factor level. There was also an anti-CD3 dose-dependent increase in the serum soluble IL-2 receptor levels. Other immune parameters monitored, including in vitro cytotoxic and proliferative responses and lymphocyte count, were similar to treatment courses with IL-2 alone. Fourteen of 26 patients examined developed human anti-murine antibodies following a single dose of anti-CD3. There were no objective antitumor responses. We conclude that in vivo treatment with anti-CD3 did not enhance T cell activity or expansion with subsequent IL-2 infusion and that the combination of anti-CD3 followed by IL-2 did not improve upon the antitumor activity previously seen with IL-2 alone.


Subject(s)
Interleukin-2/adverse effects , Muromonab-CD3/adverse effects , Neoplasms/immunology , Neoplasms/therapy , Antibody-Dependent Cell Cytotoxicity , Biomarkers/blood , Dose-Response Relationship, Drug , Dyspnea , Humans , Hypotension , Infusions, Intravenous , Interleukin-2/administration & dosage , Interleukin-2/pharmacokinetics , Lymphocyte Activation , Muromonab-CD3/administration & dosage , Muromonab-CD3/pharmacokinetics , Neoplasms/blood , Receptors, Interleukin-2/blood , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...