Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Ann Oncol ; 27(10): 1928-40, 2016 10.
Article in English | MEDLINE | ID: mdl-27672108

ABSTRACT

BACKGROUND: To evaluate the safety, tolerability, pharmacokinetics, and maximum tolerated dose (MTD) of copanlisib, a phosphatidylinositol 3-kinase inhibitor, in patients with advanced solid tumors or non-Hodgkin's lymphoma (NHL). PATIENTS AND METHODS: Phase I dose-escalation study including patients with advanced solid tumors or NHL, and a cohort of patients with type 2 diabetes mellitus. Patients received three weekly intravenous infusions of copanlisib per 28-day cycle over the dose range 0.1-1.2 mg/kg. Plasma copanlisib levels were analyzed for pharmacokinetics. Biomarker analysis included PIK3CA, KRAS, BRAF, and PTEN mutational status and PTEN immunohistochemistry. Whole-body [(18)F]-fluorodeoxyglucose positron emission tomography ((18)FDG-PET) was carried out at baseline and following the first dose to assess early pharmacodynamic effects. Plasma glucose and insulin levels were evaluated serially. RESULTS: Fifty-seven patients received treatment. The MTD was 0.8 mg/kg copanlisib. The most frequent treatment-related adverse events were nausea and transient hyperglycemia. Copanlisib exposure was dose-proportional with no accumulation; peak exposure positively correlated with transient hyperglycemia post-infusion. Sixteen of 20 patients treated at the MTD had reduced (18)FDG-PET uptake; 7 (33%) had a reduction >25%. One patient achieved a complete response (CR; endometrial carcinoma exhibiting both PIK3CA and PTEN mutations and complete PTEN loss) and two had a partial response (PR; both metastatic breast cancer). Among the nine NHL patients, all six with follicular lymphoma (FL) responded (one CR and five PRs) and one patient with diffuse large B-cell lymphoma had a PR by investigator assessment; two patients with FL who achieved CR (per post hoc independent radiologic review) were on treatment >3 years. CONCLUSION: Copanlisib, dosed intermittently on days 1, 8, and 15 of a 28-day cycle, was well tolerated and the MTD was determined to be 0.8 mg/kg. Copanlisib exhibited dose-proportional pharmacokinetics and promising anti-tumor activity, particularly in patients with NHL. CLINICALTRIALSGOV: NCT00962611; https://clinicaltrials.gov/ct2/show/NCT00962611.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Enzyme Inhibitors/administration & dosage , Lymphoma, Non-Hodgkin/drug therapy , Neoplasms/drug therapy , Pyrimidines/administration & dosage , Quinazolines/administration & dosage , Administration, Intravenous , Adult , Aged , Class I Phosphatidylinositol 3-Kinases/genetics , Dose-Response Relationship, Drug , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/pharmacokinetics , Female , Humans , Lymphoma, Non-Hodgkin/enzymology , Lymphoma, Non-Hodgkin/pathology , Male , Middle Aged , Neoplasm Staging , Neoplasms/enzymology , Neoplasms/pathology , Pyrimidines/adverse effects , Pyrimidines/pharmacokinetics , Quinazolines/adverse effects , Quinazolines/pharmacokinetics
2.
J Mol Med (Berl) ; 78(12): 673-83, 2001.
Article in English | MEDLINE | ID: mdl-11434720

ABSTRACT

Tolerance in vivo and its in vitro counterpart, anergy, are defined as the state in which helper T lymphocytes are alive but incapable of producing IL-2 and expanding in response to optimal antigenic stimulation. Anergy is induced when the T cell receptor (TCR) is engaged by antigen in the absence of costimulation or IL-2. This leads to unique intracellular signaling events that stand in contrast to those triggered by coligation of the TCR and costimulatory receptors. Specifically, anergy is characterized by lack of activation of lck, ZAP 70, Ras, ERK, JNK, AP-1, and NF-AT. In contrast, anergizing stimuli appear to activate the protein tyrosine kinase fyn, increase intracellular calcium levels, and activate Rap1. Moreover, anergizing TCR signals result in increased intracellular concentrations of the second messenger cAMP. This second messenger upregulates the cyclin-dependent kinase (cdk) inhibitor p27kip1, sequestering cyclin D2-cdk4, and cyclin E/cdk2 complexes and preventing progression of T cells through the G1 restriction point of the cell cycle. In contrast, costimulation through CD28 prevents p27kip1 accumulation by decreasing the levels of intracellular cAMP and promotes p27kip1 down-regulation due to direct degradation of the protein via the ubiquitin-proteasome pathway. Subsequent autocrine action of IL-2 leads to further degradation of p27kip1 and entry into S phase. Understanding the biochemical and molecular basis of T cell anergy will allow the development of new assays to evaluate the immune status of patients in a variety of clinical settings in which tolerance has an important role, including cancer, autoimmune diseases, and organ transplantation. Precise understanding of these biochemical and molecular events is necessary in order to develop novel treatment strategies against cancer. One of the mechanisms by which tumors down-regulate the immune system is through the anergizing inactivation of helper T lymphocytes, resulting in the absence of T cell help to tumor-specific CTLs. Although T-cells specific for tumor associated antigens are detected in cancer patients they often are unresponsive. Reversal of the defects that block the cell cycle progression is mandatory for clonal expansion of tumor specific T cells during the administration of tumor vaccines. Reversal of the anergic state of tumor specific T cells is also critical for the sufficient expansion of such T cells ex vivo for adoptive immunotherapy. On the other hand, understanding the molecular mechanisms of anergy will greatly improve our ability to design novel clinical therapeutic approaches to induce antigen-specific tolerance and prevent graft rejection and graft-versus-host disease. Such treatment approaches will allow transplantation of bone marrow and solid organs between individuals with increasing HLA disparity and therefore expand the donor pool, enable reduction in the need for nonspecific immunosuppression, minimize the toxicity of chemotherapy, and reduce the risk of opportunistic infections.


Subject(s)
Clonal Anergy/immunology , Neoplasms/physiopathology , Neoplasms/therapy , T-Lymphocytes, Helper-Inducer/immunology , Animals , Cell Cycle , Cell Cycle Proteins/metabolism , Cyclic AMP/metabolism , Cyclin-Dependent Kinase Inhibitor p27 , Humans , Immune Tolerance , Neoplasms/immunology , Signal Transduction , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/metabolism , Tumor Suppressor Proteins/metabolism
3.
Br J Cancer ; 93(1): 54-9, 2005 Jul 11.
Article in English | MEDLINE | ID: mdl-15986034

ABSTRACT

The goal of this phase I study was to develop a novel schedule using oral etoposide and infusional topotecan as a continually alternating schedule with potentially optimal reciprocal induction of the nontarget topoisomerase. The initial etoposide dose was 15 mg m(-2) b.i.d. days (D)1-5 weeks 1,3,5,7,9 and 11, escalated 5 mg per dose per dose level (DL). Topotecan in weeks 2,4,6,8,10 and 12 was administered by 96 h infusion at an initial dose of 0.2 mg m(-2) day(-1) with a dose escalation of 0.1, then at 0.05 mg m(-2) day(-1). Eligibility criteria required no organ dysfunction. Two dose reductions or delays were allowed. A total of 36 patients with a median age of 57 (22-78) years, received a median 8 (2-19) weeks of chemotherapy. At DL 6, dose-limiting toxicities consisted of grade 3 nausea, vomiting and intolerable fatigue. Three patients developed a line-related thrombosis or infection and one subsequently developed AML. There was no febrile neutropenia. There were six radiologically confirmed responses (18%) and 56% of patients demonstrated a response or stable disease, typically with only modest toxicity. Oral etoposide 35 mg m(-2) b.i.d. D1-5 and 1.8 mg m(-2) 96 h (total dose) infusional topotecan D8-11 can be administered on an alternating continual weekly schedule for at least 12 weeks, with promising clinical activity.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoplasms/drug therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Drug Administration Schedule , Etoposide/administration & dosage , Humans , Middle Aged , Topotecan/administration & dosage
4.
Clin Immunol Immunopathol ; 69(2): 223-33, 1993 Nov.
Article in English | MEDLINE | ID: mdl-8403560

ABSTRACT

Rat adenocarcinoma 13762 was adapted to continuous growth in culture and used in a variety of experiments to investigate the immune response to inoculation of animals with replication-defective tumor cells. The results demonstrate that 13762 cells express tumor-specific tumor rejection antigens that elicit protective immunity to tumorigenic challenge. By several criteria there is no apparent humoral component of the anti-tumor immunity; however, anti-tumor immunity is characterized by nylon-wool nonadherent spleen T cells. Anti-tumor T cells demonstrate tumoricidal activity in local adoptive transfer assays and are not found in spleens of naive animals or animals immunized against either nontumorigenic Rat 1 cells or a syngeneic fibrosarcoma. Despite the expression of tumor rejection antigens 13762 tumor, and the demonstrable ability of injection of irradiated tumor to induce anti-tumor immunity, tumors elicited in unimmunized syngeneic animals grow progressively. The reasons for growth of antigenic tumor are unknown but are shown not to be due to defective antigen expression in 13762 tumor since, in addition to being able to elicit T cell immune response in immunized animals, 13762 tumor expresses MHC Class I molecules and can be a target for allogeneic T cell recognition in vitro. These data suggest that in tumor-bearing animals an effective anti-tumor immune response is either not initiated or down-regulated. Since animals bearing 13762 tumors can be immunized against an unrelated syngeneic sarcoma, can produce humoral responses to several protein antigens, and can produce delayed type hypersensitivity response against dinitrofluorobenzene, the immune response to 13762-induced tumors appears specifically suppressed. In support of this contention, 13762 cells express high levels of transforming growth factor beta 1 in vitro which is postulated to impact upon the nascent anti-tumor immune response.


Subject(s)
Adenocarcinoma/immunology , Antigens, Neoplasm/biosynthesis , Immune Tolerance , Mammary Neoplasms, Experimental/immunology , Adjuvants, Immunologic , Animals , Antibodies, Neoplasm/biosynthesis , Dinitrofluorobenzene/immunology , Enzyme-Linked Immunosorbent Assay , Fibrosarcoma/immunology , Flow Cytometry , Hemagglutination Tests , Hemocyanins/immunology , Histocompatibility Antigens Class I/biosynthesis , Hypersensitivity, Delayed/chemically induced , Immunotherapy, Adoptive , Interferon-gamma/pharmacology , Mammary Neoplasms, Experimental/prevention & control , Neoplasm Transplantation , Rats , Rats, Inbred F344 , T-Lymphocytes/immunology , Time Factors , Transforming Growth Factor beta/biosynthesis , Tumor Cells, Cultured
5.
Int J Cancer ; 63(4): 576-83, 1995 Nov 15.
Article in English | MEDLINE | ID: mdl-7591269

ABSTRACT

To study the effect of a transforming allele of the tumor suppressor p53 upon the anti-tumor immune response, antigenic L-929 cells were transfected with the dominant-negative valine135 mutant of murine p53. Several p53val135-expressing transfectants formed non-regressing tumors in immunocompetent hosts. The growth rates of tumorigenic and non-tumorigenic clones were equivalent in vitro in sublethally irradiated C3H/HeN mice and in nude mice. Tumorigenic and non-tumorigenic p53val135-expressing L-929 clones expressed equivalent levels of cell surface class I major histocompatibility complex (MHC) glycoproteins. Immunization with a tumorigenic Lp53val135 clone protected mice from subsequent challenge and primed MHC class I-restricted cytotoxic T-lymphocytic precursors. Secretion of an immunosuppressive cytokine, transforming growth factor beta-1 and sensitivity to tumor necrosis factor-alpha were equivalent from tumorigenic and non-tumorigenic cell lines. These data suggest that expression of a transforming allele of p53 can allow L-929 cells to escape the host immune system.


Subject(s)
Alleles , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Genes, Dominant , Genes, p53 , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics , Animals , Histocompatibility Antigens Class I/biosynthesis , Immunocompetence , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Nude , Skin Neoplasms/genetics , T-Lymphocytes, Cytotoxic/immunology , Transfection , Transforming Growth Factor beta/metabolism , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/pharmacology , Tumor Suppressor Protein p53/immunology
6.
Int J Cancer ; 61(6): 887-94, 1995 Jun 09.
Article in English | MEDLINE | ID: mdl-7540599

ABSTRACT

To study the immune response against oncogene-transformed tumors, C3H/HcN mouse embryo fibroblasts (MEF) were transfected with an activated allele of the H-ras proto-oncogene VaII2 and a dominant-negative allele of the murine p53 tumor suppressor gene VaII35. Transformed cell lines were derived and found to be tumorigenic in syngeneic mice. Immunization with irradiated p53 + ras-transformed MEF, but not primary MEF or unrelated syngeneic cells, protected mice from subsequent challenge with live tumor cells. The role of different immune cell subsets in the effector phase of anti-tumor immunity induced by immunization with p53 + ras-transformed MEF was investigated by in vivo antibody depletion experiments. Immunized mice depleted of CD8+ T, NK or B cells were resistant, but depletion of CD4+ T cells rendered mice susceptible to tumorigenic challenge. In contrast to the tumor-specific immune responses mounted against most chemically or UV-induced tumors, a series of independently derived p53 + ras-transformed MEF were cross-reactive in tumor rejection assays. In addition, immunization with C3H-derived L-929 cell lines expressing single gene products H-ras or p53 did not protect mice against tumorigenic challenge with p53 + ras-transformed tumors. However, MEF transformed by expression of either H-ras or p53 were cross-protective in vivo. Our data suggest that the p53 + ras-transformed MEF share tumor rejection antigens which are also induced by single gene transformation of the parental primary cell but are not the products of oncogenic ras or p53 protein.


Subject(s)
Antigens, Neoplasm/biosynthesis , Cell Transformation, Neoplastic/immunology , Epitopes/biosynthesis , Fibroblasts/immunology , Animals , Antigens, Neoplasm/immunology , Cell Line, Transformed , Cell Transformation, Neoplastic/genetics , Cross Reactions , Embryo, Mammalian/cytology , Epitopes/immunology , Female , Fibroblasts/pathology , Genes, p53 , Genes, ras , Immunologic Surveillance , Mice , Mice, Inbred C3H , Mice, Inbred Strains , Transfection
7.
J Immunol ; 164(1): 144-51, 2000 Jan 01.
Article in English | MEDLINE | ID: mdl-10605005

ABSTRACT

In the presence of TCR ligation by Ag, CD28 pathway mediates the most potent costimulatory signal for T cell activation, cytokine secretion, and T cell expansion. Although CD28 costimulation promotes T cell expansion due to IL-2 secretion and subsequent signaling via the IL-2 receptor, recent studies indicate that the dramatic T cell expansion mediated through the unopposed CD28 stimulation in CTLA4-deficient mice is IL-2 independent. Therefore, we sought to dissect the effects of CD28 and IL-2 receptor pathways on cell cycle progression and determine the molecular mechanisms by which the CD28 pathway regulates T cell expansion. Here we show that CD28 costimulation directly regulates T cell cycle entry and progression through the G1 phase in an IL-2-independent manner resulting in activation of cyclin D2-associated cdk4/cdk6 and cyclin E-associated cdk2. Subsequent progression into the S phase is mediated via both IL-2-dependent and IL-2-independent mechanisms and, although in the absence of IL-2 the majority of T cells are arrested at the G1/S transition, a significant fraction of them progresses into the S phase. The key regulatory mechanism for the activation of cyclin-cdk complexes and cell cycle progression is the down-regulation of p27kip1 cdk inhibitor, which is mediated at the posttranscriptional level by its ubiquitin-dependent degradation in the proteasome pathway. Therefore, CD28 costimulation mediates T cell expansion in an IL-2-independent and IL-2 dependent manner and regulates cell cycle progression at two distinct points: at the early G1 phase and at the G1/S transition.


Subject(s)
CD28 Antigens/physiology , Cell Cycle Proteins , Cell Cycle/immunology , Interleukin-2/physiology , Lymphocyte Activation , Signal Transduction/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Tumor Suppressor Proteins , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/antagonists & inhibitors , Cysteine Endopeptidases/metabolism , Down-Regulation/genetics , Down-Regulation/immunology , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , G1 Phase/immunology , Humans , Microtubule-Associated Proteins/antagonists & inhibitors , Microtubule-Associated Proteins/biosynthesis , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/physiology , Multienzyme Complexes/metabolism , Phosphorylation , Proteasome Endopeptidase Complex , Protein Processing, Post-Translational/genetics , Protein Processing, Post-Translational/immunology , Retinoblastoma Protein/metabolism , S Phase/immunology , Signal Transduction/genetics , T-Lymphocytes/enzymology , T-Lymphocytes/metabolism , Ubiquitins/metabolism
8.
Oncologist ; 5(4): 336-44, 2000.
Article in English | MEDLINE | ID: mdl-10965002

ABSTRACT

Shortly before his death in 1995, Kenneth B. Schwartz, a cancer patient at Massachusetts General Hospital (MGH), founded The Kenneth B. Schwartz Center at MGH. The Schwartz Center is a nonprofit organization dedicated to supporting and advancing compassionate health care delivery, which provides hope to the patient, support to caregivers, and encourages the healing process. The center sponsors the Schwartz Center Rounds, a monthly multidisciplinary forum where caregivers reflect on important psychosocial issues faced by patients, their families, and their caregivers, and gain insight and support from fellow staff members. Psychosocial issues profoundly affect patients with cancer. Of the many complexities that make up the psychosocial dynamic, perhaps the medical profession is most uncomfortable with sexuality. Many elements of sexual behavior remain high-profile taboos. A number of diseases and treatments significantly affect sexual function. Male and female sexuality were discussed in two separate rounds with an emphasis on how to begin a dialogue about sexuality without jeopardizing other aspects of the relationship with patients. Three cases were presented. A patient with prostate cancer considering treatment options for early-stage disease and two patients with gynecologic malignancies; one with a colostomy following cytoreductive surgery for ovarian cancer and the other with a failed vaginal reconstruction for recurrent squamous cell carcinoma of the vagina. Staff discussed the wide diversity of response to sexual dysfunction and the difficulties that patients face. A sensitive and informed approach to discussing sexuality can provide effective support. The elements of successful dialogue are presented in the PLISSIT model.


Subject(s)
Neoplasms/psychology , Physician-Patient Relations , Sexual Dysfunction, Physiological/etiology , Sexuality , Adult , Communication , Female , Humans , Interprofessional Relations , Male , Neoplasms/complications , Neoplasms/therapy , Quality of Life
SELECTION OF CITATIONS
SEARCH DETAIL