Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 117
Filter
1.
Ann Oncol ; 30(4): 589-596, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30689736

ABSTRACT

BACKGROUND: With increasing anti-PD-1 therapy use in patients with melanoma and other tumor types, there is interest in developing early on-treatment biomarkers that correlate with long-term patient outcome. An understanding of the pathologic features of immune-mediated tumor regression is key in this endeavor. MATERIALS AND METHODS: Histologic features of immune-related pathologic response (irPR) following anti-PD-1 therapy were identified on hematoxylin and eosin (H&E)-stained slides in a discovery cohort of pre- and on-treatment specimens from n = 16 patients with advanced melanoma. These features were used to generate an irPR score [from 0 = no irPR features to 3 = major pathologic response on biopsy (MPRbx, ≤10% residual viable tumor)]. This scoring system was then tested for an association with objective response by RECIST1.1 and overall survival in a prospectively collected validation cohort of pre- and on-treatment biopsies (n = 51 on-treatment at 4-week timepoint) from melanoma patients enrolled on the nivolumab monotherapy arm of CA209-038 (NCT01621490). RESULTS: Specimens from responders in the discovery cohort had features of immune-activation (moderate-high TIL densities, plasma cells) and wound-healing/tissue repair (neovascularization, proliferative fibrosis) compared to nonresponders, (P ≤ 0.021, for each feature). In the validation cohort, increasing irPR score associated with objective response (P = 0.009) and MPRbx associated with increased overall survival (n = 51; HR 0.13; 95%CI, 0.054-0.31, P = 0.015). Neither tumoral necrosis nor pretreatment histologic features were associated with response. Eight of 16 (50%) of patients with stable disease showed irPR features, two of which were MPRbx, indicating a disconnect between pathologic and radiographic features at the 4-week on-therapy timepoint for some patients. CONCLUSIONS: Features of immune-mediated tumor regression on routine H&E-stained biopsy slides from patients with advanced melanoma correlate with objective response to anti-PD-1 and overall survival. An on-therapy biopsy may be particularly clinically useful for informing treatment decisions in patients with radiographic stable disease. This approach is inexpensive, straightforward, and widely available.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Biomarkers, Tumor/analysis , Melanoma/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Skin Neoplasms/drug therapy , Skin/pathology , Adult , Aged , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents, Immunological/pharmacology , Biopsy , Female , Humans , Ipilimumab/pharmacology , Ipilimumab/therapeutic use , Kaplan-Meier Estimate , Male , Melanoma/immunology , Melanoma/mortality , Melanoma/pathology , Middle Aged , Neoplasm, Residual , Nivolumab/pharmacology , Nivolumab/therapeutic use , Programmed Cell Death 1 Receptor/immunology , Prospective Studies , Response Evaluation Criteria in Solid Tumors , Skin/drug effects , Skin Neoplasms/immunology , Skin Neoplasms/mortality , Skin Neoplasms/pathology
2.
J Clin Oncol ; 23(24): 5542-51, 2005 Aug 20.
Article in English | MEDLINE | ID: mdl-16110015

ABSTRACT

PURPOSE: This randomized, controlled, multicenter, open-label, phase III study compared docetaxel versus paclitaxel in patients with advanced breast cancer that had progressed after an anthracycline-containing chemotherapy regimen. PATIENTS AND METHODS: Patients (n = 449) were randomly assigned to receive either docetaxel 100 mg/m2 (n = 225) or paclitaxel 175 mg/m2 (n = 224) on day 1, every 21 days until tumor progression, unacceptable toxicity, or withdrawal of consent. RESULTS: In the intent-to-treat population, both the median overall survival (OS, 15.4 v 12.7 months; hazard ratio [HR], 1.41; 95% CI, 1.15 to 1.73; P = .03) and the median time to progression (TTP, 5.7 months v 3.6 months; HR, 1.64; 95% CI, 1.33 to 2.02; P < .0001) for docetaxel were significantly longer than for paclitaxel, and the overall response rate (ORR, 32% v 25%; P = .10) was higher for docetaxel. These results were confirmed by multivariate analyses. The incidence of treatment-related hematologic and nonhematologic toxicities was greater for docetaxel than for paclitaxel; however, quality-of-life scores were not statistically different between treatment groups over time. CONCLUSION: Docetaxel was superior to paclitaxel in terms of OS and TTP. ORR was higher for docetaxel. Hematologic and nonhematologic toxicities occurred more frequently in the docetaxel group. The global quality-of-life scores were similar for both agents over time.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents, Phytogenic/therapeutic use , Breast Neoplasms/drug therapy , Paclitaxel/therapeutic use , Taxoids/therapeutic use , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Breast Neoplasms/pathology , Chi-Square Distribution , Disease Progression , Docetaxel , Female , Humans , Infusions, Intravenous , Logistic Models , Middle Aged , Proportional Hazards Models , Quality of Life , Treatment Outcome
3.
Dev Biol (Basel) ; 116: 93-107; discussion 133-43, 2004.
Article in English | MEDLINE | ID: mdl-15603186

ABSTRACT

From our way of thinking the problem facing vaccine strategies for cancer is not that we do not have "enough" tumour antigens. The problem is we cannot induce an immune response that is sufficient to mediate tumour regression. The normal "checks and balances" found in the body prevent the sustained expansion and subsequent persistence of immune killer cells. If vaccine strategies are going to become effective treatments for cancer patients, they will need to overcome this substantial roadblock. Recent developments in immunology have provided insights into the mechanisms that regulate the expansion and persistence of T cells. This has allowed investigators to reinterpret decades-old observations suggesting that chemotherapy administered before vaccination often led to a stronger immune response. This manuscript will review experiments that offer an explanation for these observations and present pre-clinical data from our laboratory that describes an innovative new approach to combining chemotherapy and vaccination. This approach is readily translatable to the clinic and is broadly applicable to any vaccine strategy for advanced cancer.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Humans
4.
Eur Surg Res ; 35(4): 346-51, 2003.
Article in English | MEDLINE | ID: mdl-12802095

ABSTRACT

The development of new therapeutic strategies for the treatment of bone metastases strongly depends on the availability of valid animal models. In this paper, we evaluate a preclinical model of bone metastases using a technique of tumor cell injection into the left heart ventricle of mice to study the efficacy of adoptive immunotherapy. Using flow cytometric analysis and histopathological and radiological examination, we investigated whether this experimental model of bony metastases using two murine cell lines of melanoma and breast cancer would be suitable for the study of adoptive immunotherapy for these diseases. We further report that anti-CD3-activated and IL-2-expanded tumor vaccine draining lymph node cells cause regression of tumor metastases, including bone metastases, following adoptive transfer to mice bearing 3-day metastases from the D5 melanoma cell line. These promising early results lead us to conclude the following: (1). this model of experimental bone metastases is suitable for the study of immunotherapy, and (2). further studies are warranted to extend these promising early findings of the therapeutic effects of adoptive immunotherapy in this animal model.


Subject(s)
Bone Neoplasms/secondary , Bone Neoplasms/therapy , Disease Models, Animal , Immunotherapy, Adoptive , Mice, Inbred C57BL , Animals , Bone Neoplasms/diagnostic imaging , Bone and Bones/diagnostic imaging , Bone and Bones/immunology , Bone and Bones/pathology , Breast Neoplasms/pathology , Cell Movement/immunology , Female , Injections, Intra-Arterial , Melanoma/secondary , Mice , Mice, Inbred BALB C , Radiography , Skin Neoplasms/pathology , T-Lymphocytes/immunology , T-Lymphocytes/transplantation
6.
Cell Immunol ; 209(2): 83-8, 2001 May 01.
Article in English | MEDLINE | ID: mdl-11446740

ABSTRACT

In this study we investigated whether T cells expressing high or low levels of CD62L were differentially susceptible to the T cell chemokine lymphotactin. We found that lymphotactin induced preferential migration of antigen-specific (CD62L(lo)) T cells over the nonspecific (CD62L(hi)) T cells in vitro and in vivo. The differing migratory abilities correlated with higher levels of mRNA encoding the lymphotactin receptor (XCR1) on the CD62L(lo) cells compared to the CD62L(hi) cells. Thus, we have identified a coupling mechanism between the activation of T cells and acquisition of new homing properties, in this case conferred by XCR1 expression. These data confirm that at least one function of lymphotactin includes mediating the recruitment of recently activated antigen-specific T cells.


Subject(s)
Chemokines, C , Chemotaxis, Leukocyte/immunology , L-Selectin/analysis , Lymphocyte Activation/immunology , Lymphokines/immunology , Membrane Proteins , Receptors, G-Protein-Coupled , Sialoglycoproteins/immunology , T-Lymphocyte Subsets/immunology , Animals , Cancer Vaccines/immunology , Mice , Mice, Inbred BALB C , Receptors, Cell Surface/biosynthesis
7.
J Immunol ; 166(12): 7370-80, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11390488

ABSTRACT

The mechanism by which tumors are rejected following the adoptive transfer of tumor-specific T cells is not well characterized. Recent work has challenged the requirement for cytotoxicity mediated by either the perforin/granzyme or Fas/Fas ligand pathway in T cell-mediated tumor regression. Many reports, including ours, suggest that tumor-specific production of IFN-gamma is critical for T cell-mediated tumor regression. However, in most of these studies the evidence to support the role for IFN-gamma is only indirect. We have directly examined the requirement for IFN-gamma using IFN-gamma knockout (GKO) mice. The results show an interesting dichotomy in the requirement for IFN-gamma: Antitumor immunity induced by active-specific immunotherapy (vaccination) required IFN-gamma, whereas adoptive immunotherapy did not. In GKO mice vaccination with the GM-CSF gene-modified B16BL6-D5 tumor (D5-G6) failed to induce protective immunity against parental D5 tumor. However, adoptive transfer of effector T cells from GKO mice cured 100% of GKO mice with established pulmonary metastases and induced long term antitumor immunity and depigmentation of skin. Furthermore, in vivo neutralization of IFN-gamma by mAb treatment or adoptive transfer into IFN-gamma receptor knockout mice failed to block the therapeutic efficacy of effector T cells generated from wild-type or perforin knockout mice. Analysis of regressing metastases revealed similar infiltrates of macrophages and granulocytes in both wild-type and GKO mice. These results indicate that in this adoptive immunotherapy model, neither a direct effect on the tumor nor an indirect effect of IFN-gamma through activation of myeloid or lymphoid cells is critical for therapeutic efficacy.


Subject(s)
Cancer Vaccines/immunology , Immunotherapy, Adoptive , Interferon-gamma/physiology , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Animals , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/therapeutic use , Cells, Cultured , Cytokines/biosynthesis , Cytotoxicity, Immunologic/genetics , Female , Immune Sera/administration & dosage , Immunohistochemistry , Immunophenotyping , Immunotherapy, Adoptive/methods , Injections, Intravenous , Injections, Subcutaneous , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/deficiency , Interferon-gamma/genetics , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymph Nodes/pathology , Lymphocyte Activation/genetics , Melanoma, Experimental/genetics , Melanoma, Experimental/mortality , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , T-Lymphocyte Subsets/transplantation , T-Lymphocytes, Cytotoxic/immunology , Th2 Cells/immunology , Th2 Cells/metabolism , Tumor Cells, Cultured/transplantation , Vitiligo/genetics , Vitiligo/immunology
8.
Cancer Invest ; 19(3): 225-33, 2001.
Article in English | MEDLINE | ID: mdl-11338878

ABSTRACT

This study evaluated mitoxantrone and paclitaxel combination chemotherapy in the treatment of patients with metastatic breast cancer. Thirty-seven patients who had developed progressive disease after prior chemotherapy were treated with mitoxantrone (14 mg/m2) and paclitaxel (150 mg/m2) every 21 days for a maximum of six cycles. The most frequent grade 3 or 4 nonhematological toxicities were fever and nausea. Grade 4 neutropenia occurred in 71% of patients. Cardiotoxicity occurred in 2 patients, both of whom had previously received doxorubicin. Objective response was achieved in 35% of patients (5% complete response and 30% partial response) and 41% had stable disease. Median time to disease progression and median survival were 6 and 12 months, respectively. The percent of patients with an objective response was not different for those who had received prior doxorubicin or had chemotherapy in the preceding 6 months. This regimen appears to be effective and well tolerated as salvage therapy and merits further evaluation.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Breast Neoplasms/pathology , Disease Progression , Disease-Free Survival , Female , Humans , Infusions, Intravenous , Middle Aged , Mitoxantrone/administration & dosage , Neoplasm Metastasis , Neutropenia/chemically induced , Paclitaxel/administration & dosage , Prognosis , Treatment Outcome
9.
Blood ; 97(7): 1942-6, 2001 Apr 01.
Article in English | MEDLINE | ID: mdl-11264156

ABSTRACT

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)


Subject(s)
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.
Cell Immunol ; 207(2): 81-8, 2001 Feb 01.
Article in English | MEDLINE | ID: mdl-11243697

ABSTRACT

We found that the murine breast cancer cell line 4T1 constitutively produced several chemokines capable of recruiting T cells. Additionally, supernatants from the tumor cell line mediated chemotaxis of T cells in a pertussis toxin-sensitive manner, indicating that these chemokines were functional. However, we also found an impaired chemotactic ability of splenic T cells in mice bearing these same tumors. The receptors for RANTES, MCP-1, and SLC were desensitized. Thus, the impaired chemotactic ability of T cells in tumor-bearing mice may explain why tumors that secrete chemokines grow progressively in a host.


Subject(s)
Neoplasms, Experimental/immunology , Receptors, Chemokine/physiology , Animals , Chemotaxis, Leukocyte , Mice , Mice, Inbred BALB C , Receptors, Chemokine/analysis , T-Lymphocytes/immunology
11.
J Immunol ; 165(8): 4246-53, 2000 Oct 15.
Article in English | MEDLINE | ID: mdl-11035058

ABSTRACT

The requirement for CD4(+) Th cells in the cross-priming of antitumor CTL is well accepted in tumor immunology. Here we report that the requirement for T cell help can be replaced by local production of GM-CSF at the vaccine site. Experiments using mice in which CD4(+) T cells were eliminated, either by Ab depletion or by gene knockout of the MHC class II beta-chain (MHC II KO), revealed that priming of therapeutic CD8(+) effector T cells following vaccination with a GM-CSF-transduced B16BL6-D5 tumor cell line occurred independently of CD4(+) T cell help. The adoptive transfer of CD8(+) effector T cells, but not CD4(+) effector T cells, led to complete regression of pulmonary metastases. Regression of pulmonary metastases did not require either host T cells or NK cells. Transfer of CD8(+) effector T cells alone could cure wild-type animals of systemic tumor; the majority of tumor-bearing mice survived long term after treatment (>100 days). In contrast, adoptive transfer of CD8(+) T cells to tumor-bearing MHC II KO mice improved survival, but eventually all MHC II KO mice succumbed to metastatic disease. WT mice cured by adoptive transfer of CD8(+) T cells were resistant to tumor challenge. Resistance was mediated by CD8(+) T cells in mice at 50 days, while both CD4(+) and CD8(+) T cells were important for protection in mice challenged 150 days following adoptive transfer. Thus, in this tumor model CD4(+) Th cells are not required for the priming phase of CD8(+) effector T cells; however, they are critical for both the complete elimination of tumor and the maintenance of a long term protective antitumor memory response in vivo.


Subject(s)
Adoptive Transfer , CD4-Positive T-Lymphocytes/immunology , Immunologic Memory , Lymphocyte Activation , Animals , Antigens, T-Independent/immunology , CD8-Positive T-Lymphocytes/transplantation , Cancer Vaccines/immunology , Cell Count , Cytotoxicity, Immunologic/genetics , Dendritic Cells/immunology , Dendritic Cells/pathology , Female , Histocompatibility Antigens Class II/genetics , Immunologic Memory/genetics , Immunophenotyping , Killer Cells, Natural/immunology , Lung Neoplasms/genetics , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymph Nodes/pathology , Lymphocyte Activation/genetics , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/prevention & control , Mice , Mice, Inbred C57BL , Mice, Knockout , Skin Pigmentation/genetics , Skin Pigmentation/immunology , Survival Analysis , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , T-Lymphocytes, Cytotoxic/immunology , Tumor Cells, Cultured/immunology , Tumor Cells, Cultured/transplantation
12.
Int J Cancer ; 87(5): 688-94, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-10925363

ABSTRACT

FVBN202 mice, which are transgenic for the rat neu gene, spontaneously develop mammary carcinomas between 6 and 7 months of age. We investigated whether these spontaneous tumors (spontaneous breast carcinoma cells, SBCC) could elicit an immune response in naive 6- to 8-week-old FVBN202 transgenic and FVBN nontransgenic mice. After s.c. injection of SBCC, the recently activated T cells, which were identified by their reduced expression of CD62L (L-selectin), were isolated from the draining lymph nodes, expanded with anti-CD3 and IL-2, and their cytokine response to tumor cells in vitro was analyzed. Tumor-vaccine draining lymph node lymphocytes (TVDLN) from transgenic mice failed to make IFN-gamma in response to the tumor cells. However, TVDLN from the nontransgenic mice exhibited a tumor-specific IFN-gamma response against the SBCC. This indicates that the SBCC are immunogenic. The lack of response in transgenic mice could not be attributed to cytokine immune deviation or T-cell signaling defects. Although transgenic mice were tolerant to their own tumors, their immune competence was established by their ability to respond in an allogeneic mixed lymphocyte reaction, to reject an allogeneic breast carcinoma cell line, and to produce a tumor-specific IFN-gamma response against a syngeneic cancer cell line. This transgenic mouse model provides the opportunity to investigate the immune response against a primary tumor cell culture rather than cell lines or clones and should prove useful for developing immunotherapies that overcome tolerance to self-tumor antigens.


Subject(s)
Genes, erbB-2 , Immune Tolerance/physiology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/immunology , Animals , Antigens, Neoplasm/immunology , CD3 Complex/immunology , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Graft Rejection/immunology , Histocompatibility Antigens Class I/biosynthesis , Histocompatibility Antigens Class I/immunology , Immune Tolerance/immunology , Interleukin-2/pharmacology , Isoantigens/immunology , L-Selectin/biosynthesis , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Neoplasm Transplantation , Rats , Receptor, ErbB-2/biosynthesis , Receptor, ErbB-2/genetics , Receptor, ErbB-2/immunology , Receptors, Antigen, T-Cell/physiology , Signal Transduction/physiology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Cells, Cultured
13.
Cancer J ; 6(3): 146-50, 2000.
Article in English | MEDLINE | ID: mdl-10882329

ABSTRACT

The purpose of this article was to evaluate the antitumor effects of a combination chemotherapy program based on ProMACE (prednisone, methotrexate, doxorubicin [Adriamycin], cyclophosphamide, etoposide) followed by a B cell-specific immunotoxin in the treatment of patients with advanced-stage indolent histology non-Hodgkin's lymphomas. We performed a prospective phase II clinical trial in a referral-based patient population. After confirmation of diagnosis and staging evaluation, 44 patients (10 small lymphocytic lymphoma, 27 follicular lymphoma, 7 mantle cell lymphoma; 30 without prior therapy, 14 previously treated) received six cycles of ProMACE-CytaBOM (cytarabine, bleomycin, vincristine [Oncovin], mechlorethamine) combination chemotherapy (with etoposide given orally daily for five days) followed by a 7-day continuous infusion of anti-B4-blocked ricin immunotoxin at 30 microg/kg/day given every 14 days for up to six cycles. A complete response was achieved in 25 of 44 patients (57%), 21 from the chemotherapy alone, 3 converted from partial to complete response with the immunotoxin, and 1 patient became a complete responder after a surgical procedure to remove an enlarged spleen that was histologically negative for lymphoma. With a median follow-up of 5 years, 14 of 25 complete responders have relapsed (56%); median remission duration was 2 years, and overall survival was 61%. Forty-two percent of the complete responders have been in continuous remission for more than 4 years. The median number of courses of immunotoxin delivered was two usually because of the development of human anti-ricin antibodies. ProMACE-CytaBOM plus anti-B4-blocked ricin does not produce durable complete remissions in the majority of patients with indolent lymphoma. However, the remissions appear quite durable (> 4 years) in about 40% of the complete responders.


Subject(s)
Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols , Immunoconjugates/therapeutic use , Immunotoxins/therapeutic use , Lymphoma/drug therapy , Ricin/therapeutic use , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bleomycin/therapeutic use , Cyclophosphamide/therapeutic use , Cytarabine/therapeutic use , Disease-Free Survival , Doxorubicin/therapeutic use , Etoposide/therapeutic use , Female , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/mortality , Lymphoma/mortality , Lymphoma, Follicular/drug therapy , Lymphoma, Follicular/mortality , Lymphoma, Mantle-Cell/drug therapy , Lymphoma, Mantle-Cell/mortality , Male , Methotrexate/therapeutic use , Middle Aged , Prednisone/therapeutic use , Time Factors , Treatment Outcome , Vincristine/therapeutic use
14.
Breast Cancer Res Treat ; 59(1): 41-8, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10752678

ABSTRACT

A number of approaches have been used to identify genes important in breast cancer. In one approach the genes already shown to be involved in other tumors, such as p53 and Her2neu, were examined. A second approach examined genes detected through genetic screening of families with a high incidence of breast cancer, for example, BRCA-1 and BRCA-2. We used a third approach, subtractive hybridization, to identify and clone genes that were preferentially expressed in breast cancer cells compared to normal mammary epithelium. Instead of analyzing breast cancer cell lines, we examined fresh human breast cancer specimens. By subtracting normal mammary epithelial cDNA from breast cancer cDNA, we were able to clone several genes overexpressed in breast cancer. Two of these genes, L19 and MLN70, were previously reported to be overexpressed in breast cancer. Three of these genes, L19, L34, and MLN70, were localized to a region on chromosome 17 where Her2/neu and BRCA-1 are found. In addition, we isolated a gene we call breast cancer associated gene-1 that was expressed almost exclusively in fresh breast cancer tissue and not in normal mammary epithelium or breast cancer cell lines. We were unable to detect expression of breast cancer associated gene-1 in cell lines from melanoma, renal cell carcinoma, lymphoma, or leukemia. The full-length sequence from two separate breast cancer specimens revealed one amino acid difference compared to the sequence from normal breast epithelial tissue. Further studies are necessary to determine whether these genes contribute to breast cancer development or can be used as therapeutic targets.


Subject(s)
Breast Neoplasms/genetics , DNA, Complementary/genetics , Gene Expression Regulation, Neoplastic , Amino Acid Sequence , Base Sequence , Breast Neoplasms/physiopathology , Female , Genes, Tumor Suppressor/genetics , Humans , Molecular Sequence Data , Tumor Cells, Cultured
15.
J Immunol ; 164(4): 2160-9, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10657670

ABSTRACT

The OX-40 receptor (OX-40R), a member of the TNFR family, is primarily expressed on activated CD4+ T lymphocytes. Engagement of the OX-40R, with either OX-40 ligand (OX-40L) or an Ab agonist, delivers a strong costimulatory signal to effector T cells. OX-40R+ T cells isolated from inflammatory lesions in the CNS of animals with experimental autoimmune encephalomyelitis are the cells that respond to autoantigen (myelin basic protein) in vivo. We identified OX-40R+ T cells within primary tumors and tumor-invaded lymph nodes of patients with cancer and hypothesized that they are the tumor-Ag-specific T cells. Therefore, we investigated whether engagement of the OX-40R in vivo during tumor priming would enhance a tumor-specific T cell response. Injection of OX-40L:Ig or anti-OX-40R in vivo during tumor priming resulted in a significant improvement in the percentage of tumor-free survivors (20-55%) in four different murine tumors derived from four separate tissues. This anti-OX-40R effect was dose dependent and accentuated tumor-specific T cell memory. The data suggest that engagement of the OX-40R in vivo augments tumor-specific priming by stimulating/expanding the natural repertoire of the host's tumor-specific CD4+ T cells. The identification of OX-40R+ T cells clustered around human tumor cells in vivo suggests that engagement of the OX-40R may be a practical approach for expanding tumor-reactive T cells and thereby a method to improve tumor immunotherapy in patients with cancer.


Subject(s)
Cancer Vaccines/immunology , Cancer Vaccines/metabolism , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Receptors, Tumor Necrosis Factor , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/metabolism , Animals , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cancer Vaccines/administration & dosage , Colorectal Neoplasms/immunology , Colorectal Neoplasms/prevention & control , Female , Humans , Ligands , Lymph Nodes/immunology , Lymph Nodes/pathology , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/prevention & control , Melanoma, Experimental/immunology , Melanoma, Experimental/prevention & control , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasm Transplantation , Receptors, Immunologic/administration & dosage , Receptors, OX40 , Sarcoma, Experimental/immunology , Sarcoma, Experimental/prevention & control , Tumor Necrosis Factor Receptor Superfamily, Member 7/administration & dosage , Tumor Necrosis Factor Receptor Superfamily, Member 7/biosynthesis
16.
J Immunol ; 163(8): 4462-72, 1999 Oct 15.
Article in English | MEDLINE | ID: mdl-10510388

ABSTRACT

The adoptive transfer of tumor-specific effector T cells can result in complete regression and cure mice with systemic melanoma, but the mechanisms responsible for regression are not well characterized. Perforin- and Fas ligand (APO-1/CD95 ligand)-mediated cytotoxicity have been proposed as mechanisms for T cell-mediated tumor destruction. To determine the role of perforin and Fas ligand (FasL) in T cell-mediated tumor regression in a murine melanoma model, B16BL6-D5 (D5), we generated D5-specific effector T cells from tumor vaccine-draining lymph nodes of wild type (wt), perforin knock out (PKO), or FasL mutant (gld) mice and treated established D5 metastases in mice with the same genotype. Effector T cells from wt, PKO and gld mice induced complete regression of pulmonary metastases and significantly prolonged survival of the treated animals regardless of their genotype. Complete tumor regression induced by PKO effector T cells was also observed in a sarcoma model (MCA-310). Furthermore, adoptive transfer of PKO and wt effector T cells provided long-term immunity to D5. Therapeutic T cells from wt, PKO, or gld mice exhibit a tumor-specific type 1 cytokine profile; they secrete IFN-gamma, but not IL-4. In these models, T cell-mediated tumor regression and long-term antitumor immunity are perforin and FasL independent.


Subject(s)
Adoptive Transfer , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Membrane Glycoproteins/physiology , T-Lymphocytes, Cytotoxic/transplantation , fas Receptor/metabolism , Adoptive Transfer/methods , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/transplantation , Cells, Cultured , Cytokines/biosynthesis , Cytotoxicity, Immunologic/genetics , Fas Ligand Protein , Female , Injections, Subcutaneous , Killer Cells, Lymphokine-Activated/immunology , Ligands , Lymphoproliferative Disorders/genetics , Lymphoproliferative Disorders/immunology , Melanoma, Experimental/genetics , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Transplantation , Perforin , Pore Forming Cytotoxic Proteins , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Th1 Cells/metabolism
17.
Int J Cancer ; 82(4): 562-8, 1999 Aug 12.
Article in English | MEDLINE | ID: mdl-10404071

ABSTRACT

We generated a monoclonal antibody (MAb), H8.98, that recognizes an antigen shared by 50% of examined renal carcinoma cell (RCC) lines and is susceptible to lysis by a Vgamma3Vdelta1(+) T-cell clone derived from RCC tumor-infiltrating lymphocytes. H8.98 inhibited Vgamma3Vdelta1(+ )T-cell clone-mediated lysis of RCC lines. It did not stain normal kidney lines, melanomas, fibroblasts, Burkitt's lymphoma or Epstein-Barr virus-transformed B-cell lines but it did stain 2 of 4 tested breast cancer lines. Through screening of a renal carcinoma cDNA library using H8.98, we isolated a cDNA clone which, upon sequencing, was found to be cytochrome b with 2 point mutations.


Subject(s)
Antigens, Neoplasm/genetics , Carcinoma, Renal Cell/immunology , Cytochrome b Group/genetics , Kidney Neoplasms/immunology , Point Mutation/genetics , Transcription, Genetic , Amino Acid Sequence , Animals , Antibody Specificity , Antigens, Neoplasm/immunology , COS Cells , Carcinoma, Renal Cell/genetics , Cytochrome b Group/immunology , Humans , Immunohistochemistry , Kidney Neoplasms/genetics , Molecular Sequence Data , T-Lymphocytes/immunology , Transfection , Tumor Cells, Cultured
18.
J Cancer Res Clin Oncol ; 125(5): 292-6, 1999.
Article in English | MEDLINE | ID: mdl-10359134

ABSTRACT

The therapeutic benefit of adding interferon alpha (IFNalpha) to established single-agent and combination chemotherapy regimens for the treatment of metastatic melanoma has not been proven. We designed the present study to estimate the response rate of IFNalpha, dacarbazine, cisplatin and tamoxifen in patients who had not been treated with systemic therapy for advanced disease. Using a schedule similar to that which had previously been shown to favor IFNalpha plus dacarbazine over dacarbazine alone, we treated patients with an "induction" regimen of IFNalpha, 15 mU m(-2) day(-1) intravenously 5 days/week for 3 weeks. Following induction, schedules of IFNalpha, 5 mU m(-2) day(-1) subcutaneously three times a week, and tamoxifen, 10 mg orally twice a day, were begun. Dacarbazine, 250 mg m(-2) day(-1) and cisplatin 33 mg m(-2) day(-1) for 3 consecutive days were repeated every 4 weeks, and subcutaneous IFNalpha and oral tamoxifen were continued until the discontinuation of chemotherapy. We treated 25 patients (18 men and 7 women, median age 52 years) and observed only 1 objective response (response rate 4%, 95% confidence interval 0.1%-20%). The toxicities of the regimen consisted of moderate myelosuppression and constitutional side-effects. On the basis of the low antitumor activity of this regimen, we do not recommend it for further study or for use as standard therapy of metastatic melanoma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Melanoma/drug therapy , Melanoma/secondary , Adolescent , Adult , Aged , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Hormonal/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cisplatin/administration & dosage , Dacarbazine/administration & dosage , Drug Administration Schedule , Female , Humans , Interferon-alpha/administration & dosage , Male , Middle Aged , Southwestern United States , Tamoxifen/administration & dosage , Treatment Outcome
19.
Blood ; 93(10): 3250-8, 1999 May 15.
Article in English | MEDLINE | ID: mdl-10233876

ABSTRACT

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

Subject(s)
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
20.
Eur J Nucl Med ; 26(4 Suppl): S50-3, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10199933

ABSTRACT

Recently, it was recognized that an immune response develops along one of two major pathways. One leads to a destructive immune response (type 1), while the alternative leads to a nondestructive immune response (type 2). Our studies in animal models suggest that therapeutic vaccines induce a tumor-specific type 1 immune response while ineffective vaccines induce a type 2 response. These results have led us to examine the immune response in sentinel lymph nodes draining tumor vaccines of patients entered onto clinical trials for melanoma, breast and renal cell cancer.


Subject(s)
Breast Neoplasms/immunology , Cancer Vaccines , Kidney Neoplasms/immunology , Lymph Nodes/immunology , Lymphatic Metastasis/immunology , Melanoma/immunology , Animals , Breast Neoplasms/therapy , Female , Humans , Kidney Neoplasms/therapy , Male , Melanoma/therapy , Mice
SELECTION OF CITATIONS
SEARCH DETAIL