Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Cancer Sci ; 109(1): 193-198, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29059492

ABSTRACT

The cyclin D-CDK4/6-INK4-Rb pathway is frequently dysregulated in cancers. Ribociclib, an orally available, selective CDK4/6 inhibitor, showed preliminary clinical activity in a phase I study in the USA and Europe for patients with solid tumors and lymphomas. The present study aimed to determine the single-agent maximum tolerated dose (MTD) and recommended dose for expansion (RDE) in Japanese patients with advanced solid tumors. Ribociclib safety, tolerability, pharmacokinetic profile, and preliminary antitumor activity were also assessed. Japanese patients with solid tumors that had progressed on prior therapies received escalating doses of single-agent ribociclib on a 3-weeks-on/1-week-off schedule. Treatment continued until the development of toxicity or disease progression. A dose escalation was planned for patients with esophageal cancer. In the dose-escalation phase, 4 patients received 400 mg ribociclib and 13 patients received 600 mg ribociclib. Four patients experienced dose-limiting toxicities, 3 of whom were in the 600 mg group. The RDE was declared to be 600 mg, and the MTD was not determined. The most frequent adverse events were hematologic and gastrointestinal. Four patients achieved stable disease at the 600 mg dose; no patients achieved complete or partial response. All patients discontinued the study, the majority due to disease progression. No patients discontinued due to adverse events. Dose escalation was not pursued due to lack of observed efficacy in esophageal cancer. At the RDE of 600 mg/d on a 3-weeks-on/1-week-off schedule, ribociclib showed acceptable safety and tolerability profiles in Japanese patients with advanced solid tumors.


Subject(s)
Aminopyridines/administration & dosage , Neoplasms/drug therapy , Purines/administration & dosage , Adult , Aminopyridines/adverse effects , Aminopyridines/pharmacology , Disease Progression , Drug Administration Schedule , Female , Humans , Japan , Male , Maximum Tolerated Dose , Middle Aged , Purines/adverse effects , Purines/pharmacology , Sample Size , Treatment Outcome
2.
Clin Cancer Res ; 26(24): 6417-6428, 2020 12 15.
Article in English | MEDLINE | ID: mdl-32998962

ABSTRACT

PURPOSE: Report results of the phase Ib dose-escalation/expansion study of triplet therapy with cyclin-dependent kinases 4 and 6 (CDK4/6) inhibitor (ribociclib), mTOR inhibitor (everolimus), and endocrine therapy (exemestane). PATIENTS AND METHODS: Postmenopausal women with hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2-), pretreated, advanced breast cancer (ABC) were enrolled. The primary objective of the dose-escalation phase was to estimate the MTD and recommended phase II dose (RP2D) of triplet therapy through evaluation of the incidence of dose-limiting toxicities. Safety, tolerability, and efficacy of the RP2D were evaluated in the dose-expansion phase in patients naïve or refractory to CDK4/6 inhibitor therapy. RESULTS: Patients (N = 116) received triplet therapy (n = 83 in the dose-escalation phase; n = 33 in the dose-expansion phase). A dose-dependent drug-drug interaction was observed for everolimus, with exposure increasing two- to fourfold in the presence of ribociclib. The RP2D was determined to be ribociclib 300 mg once daily, 3 weeks on/1 week off in a 4-week cycle, plus everolimus 2.5 mg once daily, plus exemestane 25 mg once daily taken with food. The safety profile was consistent with the known profiles of the combination partners, and preliminary evidence of antitumor activity was observed. Higher ESR1 gene expression trended with better treatment response to triplet therapy; higher gene expression of MAPK pathway genes trended with worse treatment response. CONCLUSIONS: Triplet therapy with endocrine therapy and mTOR and CDK4/6 inhibition provides clinical benefit and an acceptable safety profile in previously treated postmenopausal women with HR+, HER2- ABC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Postmenopause , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Aminopyridines/administration & dosage , Androstadienes/administration & dosage , Biomarkers, Tumor , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Dose-Response Relationship, Drug , Everolimus/administration & dosage , Female , Follow-Up Studies , Humans , Middle Aged , Prognosis , Purines/administration & dosage
3.
Nat Rev Cancer ; 17(5): 286-301, 2017 05.
Article in English | MEDLINE | ID: mdl-28338065

ABSTRACT

Over the past 25 years, research in cancer therapeutics has largely focused on two distinct lines of enquiry. In one approach, efforts to understand the underlying cell-autonomous, genetic drivers of tumorigenesis have led to the development of clinically important targeted agents that result in profound, but often not durable, tumour responses in genetically defined patient populations. In the second parallel approach, exploration of the mechanisms of protective tumour immunity has provided several therapeutic strategies - most notably the 'immune checkpoint' antibodies that reverse the negative regulators of T cell function - that accomplish durable clinical responses in subsets of patients with various tumour types. The integration of these potentially complementary research fields provides new opportunities to improve cancer treatments. Targeted and immune-based therapies have already transformed the standard-of-care for several malignancies. However, additional insights into the effects of targeted therapies, along with conventional chemotherapy and radiation therapy, on the induction of antitumour immunity will help to advance the design of combination strategies that increase the rate of complete and durable clinical response in patients.


Subject(s)
Immunotherapy/trends , Molecular Targeted Therapy/trends , Neoplasms/therapy , Animals , Combined Modality Therapy , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/immunology
4.
Hum Gene Ther ; 17(8): 798-806, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16942440

ABSTRACT

Combining chemotherapy and immunotherapy is problematic because chemotherapy can ablate the immune responses initiated by modulators of the immune system. We hypothesized that protection of immunocompetent cells from the toxic effects of chemotherapy, using drug resistance gene therapy strategies, would allow the combined use of chemotherapy and immunotherapy. In wild-type mice, the antitumor effectiveness of an immunotherapy regimen employing an agonistic anti-CD137 antibody is diminished with escalating doses of the antifolate trimetrexate (TMTX). Using retroviral gene transfer of a mutant form of dihydrofolate reductase (L22Y-DHFR), hematopoietic stem cells were genetically engineered to withstand the toxic effects of TMTX. Mice transplanted with L22Y-DHFR-modified bone marrow were then challenged with AG104 sarcoma cells and treated with TMTX only, anti-CD137 only, or a combination of chemotherapy and immunotherapy. Although tumor burden was transiently decreased during TMTX administration, no mice treated with TMTX alone survived the tumor challenge, whereas approximately 40% of transplanted mice treated with anti-CD137 alone survived. However, 100% of mice survived with complete tumor regression after transplantation with L22Y-DHFR-transduced bone marrow followed by combined treatment with TMTX and anti-CD137. In addition, adoptive transfer of splenocytes from cured mice extended the survival of tumor- bearing animals by approximately 3 weeks compared with controls. Therefore, protection of the hematopoietic system can allow for the combined administration of chemotherapy and immunotherapy, which results in complete tumor clearance.


Subject(s)
Antigens, CD , Bone Marrow Transplantation , Drug Resistance/genetics , Genetic Therapy , Hematopoietic Stem Cells/enzymology , Neoplasms, Experimental/therapy , Point Mutation , Receptors, Nerve Growth Factor , Receptors, Tumor Necrosis Factor , Sarcoma/therapy , Tetrahydrofolate Dehydrogenase/genetics , Adoptive Transfer/methods , Animals , Antibodies/immunology , Antibodies/pharmacology , Antigens, CD/immunology , Antimetabolites, Antineoplastic/administration & dosage , Combined Modality Therapy/methods , Drug Resistance/drug effects , Drug Resistance/immunology , Genetic Therapy/methods , Humans , Mice , Neoplasms, Experimental/enzymology , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Receptors, Nerve Growth Factor/immunology , Receptors, Tumor Necrosis Factor/immunology , Sarcoma/enzymology , Sarcoma/genetics , Sarcoma/immunology , Tetrahydrofolate Dehydrogenase/immunology , Transplantation, Homologous , Trimetrexate/pharmacology , Tumor Necrosis Factor Receptor Superfamily, Member 9
5.
Eur J Cancer ; 67: 1-10, 2016 11.
Article in English | MEDLINE | ID: mdl-27591414

ABSTRACT

Over the past decade, our understanding of cancer immunotherapy has evolved from assessing peripheral responses in the blood to monitoring changes in the tumour microenvironment. Both preclinical and clinical experience has taught us that modulation of the tumour microenvironment has significant implications to generating robust antitumour immunity. Clinical benefit has been well documented to correlate with a tumour microenvironment that contains a dense infiltration of CD8+CD45RO+ T effectors and a high ratio of CD8+ T cells to FoxP3+ regulatory T cells (Tregs). In preclinical tumour models, modulation of the Glucocorticoid induced TNF receptor (GITR)/GITR ligand (GITRL) axis suggests this pathway may provide the desired biological outcome of inhibiting Treg function while activating CD8+ T effector cells. This review will focus on the scientific rationale and considerations for the therapeutic targeting of GITR for cancer immunotherapy and will discuss possible combination strategies to enhance clinical benefit.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Glucocorticoid-Induced TNFR-Related Protein/agonists , Immunotherapy/methods , Neoplasms/drug therapy , Animals , Antibodies, Monoclonal/immunology , CTLA-4 Antigen/antagonists & inhibitors , Colonic Neoplasms , Disease Models, Animal , Drug Therapy, Combination , Glucocorticoid-Induced TNFR-Related Protein/immunology , Humans , Melanoma, Experimental , Molecular Targeted Therapy , Neoplasms/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Receptors, Fc/immunology , T-Lymphocytes, Regulatory/immunology
6.
J Clin Oncol ; 27(23): 3822-9, 2009 Aug 10.
Article in English | MEDLINE | ID: mdl-19581539

ABSTRACT

PURPOSE: Temsirolimus, a specific inhibitor of the mammalian target of rapamycin kinase, has shown clinical activity in mantle cell lymphoma (MCL). We evaluated two dose regimens of temsirolimus in comparison with investigator's choice single-agent therapy in relapsed or refractory disease. PATIENTS AND METHODS: In this multicenter, open-label, phase III study, 162 patients with relapsed or refractory MCL were randomly assigned (1:1:1) to receive one of two temsirolimus regimens: 175 mg weekly for 3 weeks followed by either 75 mg (175/75-mg) or 25 mg (175/25-mg) weekly, or investigator's choice therapy from prospectively approved options. The primary end point was progression-free survival (PFS) by independent assessment. RESULTS: Median PFS was 4.8, 3.4, and 1.9 months for the temsirolimus 175/75-mg, 175/25-mg, and investigator's choice groups, respectively. Patients treated with temsirolimus 175/75-mg had significantly longer PFS than those treated with investigator's choice therapy (P = .0009; hazard ratio = 0.44); those treated with temsirolimus 175/25-mg showed a trend toward longer PFS (P = .0618; hazard ratio = 0.65). Objective response rate was significantly higher in the 175/75-mg group (22%) compared with the investigator's choice group (2%; P = .0019). Median overall survival for the temsirolimus 175/75-mg group and the investigator's choice group was 12.8 months and 9.7 months, respectively (P = .3519). The most frequent grade 3 or 4 adverse events in the temsirolimus groups were thrombocytopenia, anemia, neutropenia, and asthenia. CONCLUSION: Temsirolimus 175 mg weekly for 3 weeks followed by 75 mg weekly significantly improved PFS and objective response rate compared with investigator's choice therapy in patients with relapsed or refractory MCL.


Subject(s)
Antineoplastic Agents/therapeutic use , Lymphoma, Mantle-Cell/drug therapy , Protein Kinase Inhibitors/therapeutic use , Protein Kinases/drug effects , Sirolimus/analogs & derivatives , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Disease-Free Survival , Drug Administration Schedule , Drug Resistance, Neoplasm , Female , Humans , Kaplan-Meier Estimate , Lymphoma, Mantle-Cell/pathology , Lymphoma, Non-Hodgkin/drug therapy , Male , Middle Aged , Neoplasm Staging , Prospective Studies , Protein Kinase Inhibitors/administration & dosage , Recurrence , Sirolimus/administration & dosage , Sirolimus/therapeutic use , TOR Serine-Threonine Kinases
7.
Curr Cancer Drug Targets ; 7(1): 55-70, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17305478

ABSTRACT

Central to the normal function of the immune system is its ability to distinguish between self and non-self since failure to do so could provoke the onset of autoimmune disease. To avoid this possibility, the immune system employs several processes that include, negative selection, peripheral tolerance, and limiting DC antigen priming of naïve T cells to the lymph nodes. Naïve T cells must receive two independent signals from these antigen-presenting cells (APC) that other cells cannot provide if they are to become productively activated. The first is antigen-specific and occurs when T cell antigen receptors encounter the appropriate antigen-MHC complex on the APC--Signal 1. A second, antigen-independent signal is delivered through a T cell costimulatory molecule that engages its APC-expressed ligands--Signal 2. In the absence of a costimulatory signal T cells typically enter a state of anergy. Furthermore, the extent to which T cell activation occurs can be held in check through specific inhibitory receptors expressed on T cells. Understanding the basic mechanisms of how T cell activation is regulated has led to the development of therapeutic approaches for targeting T cell costimulatory and inhibitory pathways for turning on, or preventing the turning off immune responses in subjects with cancer. In this review we will discuss several T cell costimulatory and inhibitory pathways known to influence the development of anti-tumor immunity and how experimental manipulation of these signaling pathways has led to the generation of protective, or curative anti-tumor immunity in mice and humans.


Subject(s)
B7-1 Antigen/physiology , CD28 Antigens/physiology , Lymphocyte Activation , Neoplasms/immunology , T-Lymphocytes/immunology , Animals , Antigen-Presenting Cells/physiology , Antigens, CD/physiology , Antigens, Differentiation/physiology , B7-2 Antigen/physiology , B7-H1 Antigen , CTLA-4 Antigen , Humans , Intercellular Signaling Peptides and Proteins/physiology , OX40 Ligand/physiology , Programmed Cell Death 1 Ligand 2 Protein , Signal Transduction , Tumor Necrosis Factor Receptor Superfamily, Member 9/physiology
8.
J Immunol ; 178(7): 4194-213, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17371976

ABSTRACT

CD137-mediated signals costimulate T cells and protect them from activation-induced apoptosis; they induce curative antitumor immunity and enhance antiviral immune responses in mice. In contrast, anti-CD137 agonistic mAbs can suppress T-dependent humoral immunity and reverse the course of established autoimmune disease. These results have provided a rationale for assessing the therapeutic potential of CD137 ligands in human clinical trials. In this study, we report that a single 200-mug injection of anti-CD137 given to otherwise naive BALB/c or C57BL/6 mice led to the development of a series of immunological anomalies. These included splenomegaly, lymphadenopathy, hepatomegaly, multifocal hepatitis, anemia, altered trafficking of B cells and CD8 T cells, loss of NK cells, and a 10-fold increase in bone marrow (BM) cells bearing the phenotype of hemopoietic stem cells. These events were dependent on CD8 T cells, TNF-alpha, IFN-gamma, and type I IFNs. BM cells up-regulated Fas, and there was a significant increase in the number of CD8+ T cells that correlated with a loss of CD19+ and Ab-secreting cells in the BM. TCR Valphabeta usage was random and polyclonal among liver-infiltrating CD8 T cells, and multifocal CD8+ T cell infiltrates were resolved upon termination of anti-CD137 treatment. Anti-CD137-treated mice developed lymphopenia, thrombocytopenia, and anemia, and had lowered levels of hemoglobin and increased numbers of reticulocytes.


Subject(s)
Cytokines/metabolism , Hematologic Diseases/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/antagonists & inhibitors , Anemia/immunology , Animals , Antibodies, Monoclonal/toxicity , Cell Movement , Cell Proliferation , Female , Hematopoiesis/immunology , Liver/immunology , Liver/pathology , Lung/immunology , Lung/pathology , Lymph Nodes/cytology , Lymphocyte Activation , Lymphocytes/immunology , Lymphopenia/immunology , Mice , Mice, Inbred Strains , Receptors, Antigen, T-Cell/immunology , Spleen/immunology , Spleen/pathology , Thrombocytopenia/immunology
9.
Biochem Biophys Res Commun ; 330(3): 768-75, 2005 May 13.
Article in English | MEDLINE | ID: mdl-15809063

ABSTRACT

Using the cationic polymer, Polybrene, and the anionic polymer, chondroitin sulfate C, we concentrated recombinant retrovirus pseudotyped with an ecotropic envelope, which is susceptible to inactivation by high-speed concentration methods. To evaluate gene marking, murine bone marrow was harvested from C3H mice, transduced with polymer-concentrated GFP virus, and transplanted into lethally irradiated recipients. Total gene marking in mice averaged 30-35% at 8 weeks post-transplant and transgene expression remained stable for over 16 weeks. Using the polymer concentration method, a second retroviral vector encoding the drug resistant variant of dihydrofolate reductase (L22Y-DHFR) was concentrated and tested. Approximately 40% of transduced murine bone marrow progenitor cells were protected against trimetrexate concentrations that completely eliminated the growth of non-modified cells. These results show that anionic and cationic polymers can be combined to rapidly concentrate viruses that are normally difficult to concentrate, and the concentrated virus efficiently transduces hematopoietic stem cells.


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Polymers/chemistry , Retroviridae/chemistry , Retroviridae/genetics , Transduction, Genetic/methods , Animals , Bone Marrow Cells/virology , Cell Line , Chondroitin Sulfates/chemistry , Hexadimethrine Bromide/chemistry , Mice , Retroviridae/physiology , Tetrahydrofolate Dehydrogenase/genetics , Tetrahydrofolate Dehydrogenase/metabolism , Transduction, Genetic/instrumentation , Transgenes/genetics
SELECTION OF CITATIONS
SEARCH DETAIL