Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 244
Filter
1.
Curr Cancer Drug Targets ; 9(8): 982-1004, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20025606

ABSTRACT

The requirements for a cell surface molecule to be suitable as an antibody-drug conjugate target are stringent. The notion that antibodies-directed toward targets on the surface of malignant cells could be used for drug delivery is not new. The history of antibody-drug conjugates has been marked by hurdles identified and overcome. Early conjugates used mouse antibodies, drugs that were either not sufficiently potent, were immunogenic (proteins) or were too toxic and linkers that were not sufficiently stable in circulation. Three main avenues have been explored using antibodies to target cytotoxic species to malignant cells, antibody-protein toxin conjugates (or antibody fragment-protein toxin fusion proteins), antibody-small molecule drug conjugates and antibody-enzyme conjugates administered along with small molecule prodrugs requiring metabolism by the conjugated enzyme to release the activate species. This review focuses on cell surface proteins that have been targeted primarily by antibody-small molecule drug conjugates and briefly discusses 34 targets being investigated. While only one antibody-drug conjugate has reached regulatory approval, there are nearly 20 of these in clinical trial. The time may have come for this technology to become a major contributor to improving treatment for cancer patients.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Immunoconjugates/pharmacology , Immunotoxins/pharmacology , Neoplasms/drug therapy , Neoplasms/immunology , Prodrugs/pharmacology , Animals , Humans , Mice
2.
Int J Hyperthermia ; 17(5): 401-14, 2001.
Article in English | MEDLINE | ID: mdl-11587078

ABSTRACT

It has previously been demonstrated that hyperthermia can activate prostaglandin synthesis and that prostaglandins are protective against hyperthermia. This study examined the use of inhibitors of prostaglandin synthesis on the response of prostate tumours to hyperthermia. The non-steroidal anti-inflammatory drugs (NSAID) ibuprofen and sulindac, known cyclooxygenase inhibitors that inhibit prostaglandin production, were effective hyperthermia sensitizers and augmented growth delay of DU-145 and PC-3 prostate tumours to combined radiation and hyperthermia treatment protocols. Pre-treatment of mice with ibuprofen and sulindac at hyperthermia sensitizing doses resulted in significant (p < 0.01) inhibition of hyperthemia-induced serum prostaglandin E2. These findings indicate that NSAID may have both sensitizing effects on prostate tumour growth and may function by inhibiting prostaglandin synthesis.


Subject(s)
Cyclooxygenase Inhibitors/therapeutic use , Hyperthermia, Induced , Ibuprofen/therapeutic use , Prostatic Neoplasms/therapy , Radiation-Sensitizing Agents/therapeutic use , Sulindac/therapeutic use , Animals , Combined Modality Therapy , Dinoprostone/antagonists & inhibitors , Dinoprostone/blood , Humans , Leukotriene B4/blood , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/radiotherapy
3.
Cancer Chemother Pharmacol ; 48(2): 151-9, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11561781

ABSTRACT

PURPOSE: This study was designed to assess the efficacy of gemcitabine plus vinorelbine using the mouse Lewis lung carcinoma model and to translate this regimen to a phase I clinical study of these two agents in patients with advanced lung cancer. MATERIALS AND METHODS: Using the mouse Lewis lung cancer model, employing growth delay and isobologram analysis, we demonstrated that gemcitabine, in combination with vinorelbine, produced additive activity with little increased toxicity over a wide range of doses. At the highest dose level studied, antagonism was observed. Based on these results, we initiated a phase IGemcitabine and vinorelbine in patients with advanced lung cancer: preclinical studies and report of a phase I trial study of this combination at the Dana Farber Cancer Institute (DFCI) in patients with untreated or once pretreated non-small-cell lung cancer (NSCLC) or once pretreated small-cell lung cancer (SCLC). Vinorelbine (given in an intravenous bolus) and gemcitabine (given in a 30-min infusion) were initially administered to patients at a dose of 15 mg/m2 and 500 mg/m2, respectively, on days 1, 8, and 15 of a 28-day cycle. Seven dose levels were subsequently explored over the course of the study. There was no intrapatient dose escalation. RESULTS: From November 1996 to March 1998, 40 patients were enrolled: 32 had NSCLC, 5 had SCLC and 3 had mixed disease (both SCLC and NSCLC). The patients were evenly divided by gender, the median age was 58 years (range 38 to 73 years), and the median ECOG performance status was 1 (range 0 to 2). All patients had normal renal and hepatic function and none had previously received gemcitabine or vinorelbine. Toxic reactions included mild to moderate fatigue, nausea, constipation, and, most significantly, neutropenia and thrombocytopenia. Phlebitis was a major problem when central venous lines were not used with 15% grade 1/2 events. The day-15 dose was held in 43% of patients at the expanded dose. No true maximum tolerated dose was reached after completion of seven dose levels. Dose level 4 (22.5 mg/m2 vinorelbine and 1,000 mg/m2 gemcitabine) was chosen for expansion and future study due to the potential increased ability of patients to receive the full doses on time. CONCLUSIONS: We conclude that this drug combination and dosage are feasible and have potential as either a front- or second-line chemotherapeutic regimen for advanced lung cancer, and phase II/III trials should be performed. However, hematologic toxicities, as found in this study, could probably be reduced with treatment on days 1 and 8 every 21 days, and current literature would suggest this to be the preferred schedule.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Lung Neoplasms/drug therapy , Adult , Aged , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/pathology , Cell Division/drug effects , Deoxycytidine/administration & dosage , Deoxycytidine/adverse effects , Deoxycytidine/analogs & derivatives , Dose-Response Relationship, Drug , Drug Synergism , Female , Humans , Lung Neoplasms/pathology , Male , Mice , Middle Aged , Vinblastine/administration & dosage , Vinblastine/adverse effects , Vinblastine/analogs & derivatives , Vinorelbine , Gemcitabine
4.
In Vivo ; 15(3): 185-93, 2001.
Article in English | MEDLINE | ID: mdl-11491013

ABSTRACT

Hepatocellular carcinoma and gastric cancer are the most prevalent tumors worldwide. Hep3B hepatocellular carcinoma and HS746T gastric cancer were used as models for these diseases in culture and in vivo. The PKC beta inhibitor 317615.2HCl was not very cytotoxic toward HS746T or Hep3B cells in culture and was, in the main, additive in cytotoxicity with cisplatin, 5-fluorouracil and gemcitabine when cell in monolayer were exposed to these agents in combination with 317615.2HCl. Treatment of nude mice bearing HS746T or Hep3B xenografts with 317615.2HCl orally twice daily resulted in a small decreased in CD31-stainable intratumoral vessels in the HS746T tumors and 60% decrease in CD31-stainable vessels in the Hep3B tumors. Somewhat larger decreases were observed in the vessel stained with CD105. As a single agent 317615.2HCl produced tumor growth delays between 6.5 and 15 days in the HS746T xenograft and between 5 and 25 days in the Hep3B xenograft over the dosage range (3 to 30 mg/kg). Sequential and simultaneous combinations with 317615.2HCl and 5-fluorouracil and gemcitabine resulted in increases in tumor growth delay on both schedules. Gemcitabine produced a 15-day tumor growth delay of the HS746T gastric carcinoma that was increased to 40 days when combined simultaneously with 317615.2HCl and to 30 days with the sequential treatment regimen. 5-Fluorouracil produced a 9-day tumor growth delay of the Hep3B hepatocellular carcinoma that increased to 31 days by simultaneous treatment with 317615.2HCl and to 43 days with the sequential treatment regimen. Treatment with the protein kinase C beta inhibitor 317615.2HCl decreased HS746T and Hep3B angiogenesis and improved treatment outcome with 5-fluorouracil and gemcitabine.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Deoxycytidine/analogs & derivatives , Isoenzymes/antagonists & inhibitors , Liver Neoplasms/drug therapy , Neovascularization, Pathologic/enzymology , Protein Kinase C/antagonists & inhibitors , Stomach Neoplasms/drug therapy , Aged , Animals , Antimetabolites, Antineoplastic/pharmacology , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/pathology , Child , Deoxycytidine/pharmacology , Enzyme Inhibitors/pharmacology , Fluorouracil/pharmacology , Humans , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Male , Mice , Mice, Nude , Neoplasm Transplantation , Neovascularization, Pathologic/pathology , Organic Chemicals , Protein Kinase C beta , Stomach Neoplasms/enzymology , Stomach Neoplasms/pathology , Tumor Cells, Cultured/transplantation , Xenograft Model Antitumor Assays , Gemcitabine
5.
Int J Hyperthermia ; 17(4): 347-56, 2001.
Article in English | MEDLINE | ID: mdl-11471985

ABSTRACT

Tumour hyperthermia, although potentially a powerful therapeutic agent and radiation sensitizer, is hindered by a number of considerations including inhomogeneous heating of deep seated tumours due to energy deposition and perfusion issues. One solution is to design hyperthermia sensitizers to amplify the effects of hyperthermia, particularly at cold spots within the tumour undergoing treatment. This study examined the use of Quercetin, a flavonoid drug shown previously to antagonize the expression of HSP72 and induce apoptosis as a sensitizer of prostate cancer growth in vivo. Quercetin dose-dependently suppressed PC-3 tumour growth in vitro and in vivo. When combined in a treatment protocol with hyperthermia, quercetin drastically inhibited tumour growth and potently amplified the effects of hyperthermia on two prostate tumour types, PC-3 and DU-145 in vivo. These experiments, thus, suggest the use of Quercetin as a hyperthermia sensitizer in the treatment of prostate carcinoma.


Subject(s)
Adenocarcinoma/therapy , Hyperthermia, Induced , Prostatic Neoplasms/therapy , Quercetin/therapeutic use , Radiation-Sensitizing Agents/therapeutic use , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Animals , Combined Modality Therapy , Humans , In Vitro Techniques , Male , Mice , Mice, Nude , Neoplasm Transplantation , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Quercetin/pharmacology , Radiation-Sensitizing Agents/pharmacology , Tumor Cells, Cultured
6.
Curr Pharm Des ; 7(13): 1259-76, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11472266

ABSTRACT

The antitumor cryptophycins are synthetic derivatives of the desipeptide cryptophycins isolated from the cyanobacterium Nostoc sp. Cryptophycin 52 that is currently in clinical trial in solid tumors, is prepared by total synthesis of four key fragments that are coupled to form the final product. The mechanism of anticancer activity of the cryptophycins has been associated with their destabilization of microtubules and with their induction of bcl-2 phosphorylation leading to apoptosis. The cryptophycins maintain activity against ovarian and breast carcinoma cells that overexpress the multidrug resistance efflux pump P-glycoprotein. Cryptophycin 52 has demonstrated a broad range of antitumor activity against both murine and human tumors. In the human MX-1 breast carcinoma xenograft cryptophycin 55 produced greater-than- additive tumor response in combination with 5-fluorouracil. In human non-small cell lung carcinoma and human small cell carcinoma xenografts, administration of the cryptophycins along with gemcitabine, cisplatin or carboplatin resulted in antitumor activity greater than either agent alone. The cryptophycins appear to be additive with fractionated radiation therapy in the human H460 non-small cell lung carcinoma. In the human HCT116 colon carcinoma, the cryptophycins resulted in a greater than additive tumor response when administered sequentially with 5-fluorouracil or irinotecan. Treatment of animals bearing intraperitoneal human OVCAR-2 ovarian carcinoma with cryptophycin 52 resulted in survival times that were greater than those achieved with docetaxel or paclitaxel. Cryptophycin 52 is currently in early clinical testing.


Subject(s)
Antineoplastic Agents/pharmacology , Depsipeptides , Lactams/pharmacology , Lactones/pharmacology , Peptides, Cyclic/pharmacology , Animals , Humans , Lactams/chemical synthesis , Lactams/therapeutic use , Lactones/chemical synthesis , Lactones/therapeutic use , Phosphorylation , Proto-Oncogene Proteins c-bcl-2/metabolism
7.
Clin Cancer Res ; 7(3): 634-40, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11297259

ABSTRACT

Although rare, the morbidity and mortality from brain tumors are significant. Chemotherapy has made only a small impact on these tumors. The human T98G glioblastoma multiforme cell line was used as a brain tumor model. The protein kinase Cbeta inhibitor 317615 x 2HCl was not highly cytotoxic toward T98G cells in culture and was additive in cytotoxicity with carmustine (BCNU). When nude mice bearing s.c. T98G tumors were treated with 317615 x 2HCl p.o. twice daily on days 14-30 after tumor cell implantation, the number of intratumoral vessels stained by CD31 was decreased to 37% of control, and the number of intratumoral vessels stained by CD105 was decreased to 50% of control. The compound 317615 x 2HCl was an active antitumor agent against s.c. growing T98G xenografts. A treatment regimen administering 317615 x 2HCl before, during, and after BCNU was compared with a treatment regimen administering 317615 x 2HCl sequentially after BCNU. In the tumor growth delay determination of the s.c. tumor, the sequential treatment regimen was more effective than the simultaneous treatment regimen. However, when the same treatments were administered to animals bearing intracranial T98G tumors, the survival of animals receiving the simultaneous treatment regimen increased from 41 days for those treated with BCNU alone to 102 days for animals treated with the combination, whereas animals receiving the sequential treatment regimen survived 74 days. Treatment with the protein kinase Cbeta inhibitor decreased T98G glioblastoma multiforme angiogenesis and improved treatment outcome with BCNU.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use , Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Enzyme Inhibitors/pharmacology , Glioblastoma/drug therapy , Isoenzymes/antagonists & inhibitors , Protein Kinase C/antagonists & inhibitors , Animals , Antineoplastic Agents, Alkylating/chemistry , Brain Neoplasms/blood supply , Carmustine/therapeutic use , Cell Survival , Dose-Response Relationship, Drug , Glioblastoma/blood supply , Humans , Inhibitory Concentration 50 , Male , Mice , Mice, Nude , Neoplasm Transplantation , Neovascularization, Pathologic , Organic Chemicals , Protein Kinase C beta , Time Factors , Tumor Cells, Cultured
8.
Cancer Chemother Pharmacol ; 47(2): 170-8, 2001.
Article in English | MEDLINE | ID: mdl-11269744

ABSTRACT

Bcl-2 has been described as a factor that can protect from apoptosis. The protective effect of Bcl-2 may be lost if the protein is phosphorylated. Bcl-2 phosphorylation can be induced by agents that affect microtubule depolymerization or prevent microtubule assembly. In 13 human tumor cell lines there was a high degree of heterogeneity in Bcl-2 protein expression. Human H460 non-small-cell lung carcinoma (NSCLC) cells expressed high levels of Bcl-2 and were selected for study. Western blot analysis for Bcl-2 phosphorylation was carried out after 4 h and 24 h of exposure to cryptophycin 52, cryptophycin 55, paclitaxel or vinblastine. Cryptophycin 52 and cryptophycin 55 were very potent inducers of Bcl-2 phosphorylation. After 4 h of exposure, Bcl-2 phosphorylation was evident with 0.05 nM cryptophycin 52, 0.25 nM cryptophycin 55, 5 nM vinblastine and 50 nM paclitaxel. The hyperphosphorylated form of Bcl-2 was evident after 24 h exposure of H460 cells to 0.25 nM cryptophycin 52 or cryptophycin 55 and 50 nM vinblastine or paclitaxel. The effects of the compounds on the cell cycle paralleled those on Bcl-2 phosphorylation. In H460 cells 90% cell killing was obtained with 0.13 nM cryptophycin 52, 0.2 nM cryptophycin 55, 20 nM paclitaxel and > 100 nM vinblastine after 24 h of exposure as determined by colony formation. In Bcl-2-negative Calu-6 NSCLC cells, 90% cell killing was obtained with 0.03 nM cryptophycin 52, 0.1 nM cryptophycin 55, 11 nM paclitaxel and 0.5 nM vinblastine using the same experimental design. Thus, cryptophycins are potent inducers of Bcl-2 phosphorylation. The cryptophycins were more potent cytotoxic agents in Bcl-2-negative Calu-6 cells than in Bcl-2-positive H460 cells indicating that pathways triggered by Bcl-2 phosphorylation are involved in cryptophycin-induced lethality.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Peptides, Cyclic/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Blotting, Western , Carcinoma, Non-Small-Cell Lung/pathology , Cell Division/drug effects , Cell Survival/drug effects , Depsipeptides , G2 Phase/drug effects , Humans , Lung Neoplasms/pathology , Mitosis/drug effects , Phosphorylation , Tumor Cells, Cultured
9.
Cancer Chemother Pharmacol ; 48(6): 473-80, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11800028

ABSTRACT

In cell culture, the compound 317615.2HCl, a potent inhibitor of VEGF-stimulated HUVEC proliferation, was not very effective against Calu-6 non-small-cell lung carcinoma cells (IC50 26 microM). Exposure to combinations of paclitaxel or carboplatin and 317615.2HCl with Calu-6 cells in culture resulted in cell survival that reflected less-than-additivity to additivity of the two agents. Administration of 317615.2HCl orally twice daily to nude mice bearing subcutaneous Calu-6 tumors resulted in a decreased number of intratumoral vessels as determined by CD31 and CD105 staining to 50% of the number in control tumors. 317615.2HCl showed antitumor activity against the Lewis lung carcinoma and increased the tumor growth delay produced by paclitaxel by 5-fold, that produced by gemcitabine by 2-fold and that produced by carboplatin by 1.7-fold. There was a decrease in the number of lung metastases in the Lewis lung carcinoma that paralleled the increased response of the primary tumor with each treatment combination. Administration of 317615.2HCl also increased the tumor growth delay produced by fractionated radiation therapy in the Lewis lung tumor. Treatment with 317615.2HCl was an effective therapy in the Calu-6 non-small-cell lung carcinoma xenograft when the compound was administered early (days 4-18) or later (days 14-30). Combination treatment regimens in which 317615.2HCl was administered along with or sequentially with paclitaxel or carboplatin were much more effective than the chemotherapeutic agents administered alone. 317615.2HCl is in early clinical testing.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents, Alkylating/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Enzyme Inhibitors/pharmacology , Isoenzymes/antagonists & inhibitors , Isoenzymes/pharmacology , Lung Neoplasms/drug therapy , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/pharmacology , Administration, Oral , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Humans , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Mice , Mice, Nude , Neoplasms, Experimental , Organic Chemicals , Protein Kinase C beta , Rats , Rats, Inbred Lew , Transplantation, Heterologous
10.
Cancer Metastasis Rev ; 20(1-2): 133-43, 2001.
Article in English | MEDLINE | ID: mdl-11831642

ABSTRACT

Malignant cells survive and thrive by expressing growth and invasion 'programs' that many normal cell types recognize and respond to in 'programmed' patterns. An early event in the molecular evolution of many malignancies loss of response to growth control by transforming growth factor-beta (TGF-beta) frequently due to mutation in the type I or type II TGF-beta receptor or a Smad protein. The malignant cells secrete TFG-beta that acts on the host to suppress antitumor immune responses, to enhance extracellular matrix production and to augment angiogenesis. These activities resemble those induced by TGF-beta during embryonic development and account in part for the 'de-differentiated' nature of malignant disease. Clinically, TGF-beta1 is often elevated in the plasma of breast cancer patients, lung cancer patients, hepatocellular carcinoma patients, and prostate cancer patients. Preclinically, several breast cancer models and prostate cancer models in vivo have demonstrated a connection between TGF-beta expression and increased tumorigenicity, increased invasion and drug resistance. In other diseases such as colon, gastric, endometrial, ovarian, and cervical cancers and gliomas and melanoma, loss of response to TGF-beta as a growth inhibitor and increased expression of TGF-beta have been associated with malignant conversion and progression. Elevated levels of TGF-beta are measurable in nude mice bearing a wide variety of human tumor xenografts; thus, these tumor models may serve as useful mimics of the human disease with respect to the TGF-beta pathway. Cancer cure may be approached by blocking several of the major normal pathways used for tumor growth and survival in combination with cytotoxic therapies.


Subject(s)
Cell Division/physiology , Neoplasms/metabolism , Transforming Growth Factor beta/physiology , Animals , Cell Differentiation/physiology , Humans , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Signal Transduction
11.
Anticancer Res ; 21(5): 3175-84, 2001.
Article in English | MEDLINE | ID: mdl-11848470

ABSTRACT

The compound 317615 x 2HCl, a selective protein kinase Cbeta inhibitor, was not very cytotoxic toward human CaKi1 renal cell carcinoma cells or human HT-29 colon carcinoma cells in monolayer culture. Isobologram analysis was used to determine additivity or synergy of the combination regimens. Exposure of CaKi1 cells to 317615 x 2HCl (10 or 100 mM) along with gemcitabine or 5-fluorouracil for 24 hours resulted in cytotoxicity that appeared to be less-than-additive to additive for the two agents. Exposure of HT-29 cells to gemcitabine along with 317615 x 2HCl (10 mM or 100 mM) resulted in a synergistic cytotoxicity while combinations with 5-fluorouracil resulted in additive to greater-than-additive cytotoxicity for the agents. After treatment of CaKi1 or HT-29 xenograft-bearing mice with 317615 x 2HCl, immunohistochemical staining for expression of endothelial specific markers, either CD31 or CD105, was used to quantify the number of intratumoral vessels in the samples. CaKi1 tumor angiogenesis was very responsive to treatment with 317615 x 2HCl such that the number of intratumoral vessels stained by CD31 or CD105 was decreased to 20% of the control. The HT-29 colon carcinoma angiogenesis was also responsive to 317615 x 2HCl, such that the number of intratumoral vessels stained by CD31 or CD105 was decreased to 40% to 50% of the controL 5-fluorouracil, cisplatin or fractionated radiation therapy was combined with treatment with 317615 x 2HCl in the simultaneous combination treatment regimen in animals bearing HT-29 colon carcinoma xenografts. The resulting tumor growth delays indicated that administration of 317615 x 2HCl increased the effects of the cytotoxic therapy. Both a simultaneous or an overlapping treatment regimen and a sequential treatment regimen were used to assess 317615 x 2HCl alone and along with fractionated radiation therapy or gemcitabine against the human CaKi1 renal cell carcinoma xenograft. The CaKi1 tumor was quite sensitive to fractionated radiation therapy and to gemcitabine and, although 317615 x 2HCl was an effective single agent in this tumor, the combination regimens did not reach additivity for the combination regimens in vivo. 317615 x 2HCl is in early clinical testing.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Carcinoma, Renal Cell/drug therapy , Colonic Neoplasms/drug therapy , Deoxycytidine/analogs & derivatives , Enzyme Inhibitors/pharmacology , Isoenzymes/antagonists & inhibitors , Kidney Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Protein Kinase C/antagonists & inhibitors , Adult , Angiogenesis Inhibitors/pharmacology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Renal Cell/blood supply , Carcinoma, Renal Cell/enzymology , Cisplatin/pharmacology , Colonic Neoplasms/blood supply , Colonic Neoplasms/enzymology , Deoxycytidine/pharmacology , Drug Synergism , Female , Fluorouracil/pharmacology , HT29 Cells/drug effects , HT29 Cells/enzymology , Humans , Kidney Neoplasms/blood supply , Kidney Neoplasms/enzymology , Male , Mice , Mice, Nude , Middle Aged , Organic Chemicals , Protein Kinase C beta , Xenograft Model Antitumor Assays , Gemcitabine
12.
Cancer Chemother Pharmacol ; 46(4): 293-304, 2000.
Article in English | MEDLINE | ID: mdl-11052627

ABSTRACT

PURPOSE: Cell cycle regulatory components are interesting targets for cancer therapy. Expression of pRb, cyclin D1, cdk4, cyclin E, cdk2, E2F1 and DP-1 was determined in MCF-7 and MDA-MB-468 breast carcinoma cells, H460 and Calu-6 non-small cell lung carcinoma cells, H82 and SW2 small cell lung carcinoma cells, HCT116 and HT29 colon carcinoma cells and LNCaP and DU-145 prostate carcinoma cells. METHODS: For Western blotting, the ratio with actin expression was used to normalize the data; all lines were run on the same gels. RESULTS: In cell culture, pRb was not detected in MB-468 and H82 was low in SW2 and DU-145 and highest in HCT116; in tumors, pRb was not detected in MB-468, H82, SW2, and DU-145 and was highest in LNCaP and Calu-6. Cyclin D1 was not detected in SW2 cells in culture, was low in MB-468 and H82, and was highest in LNCaP and H460; in tumors, cyclin D1 was low in MB-468, H460, SW2 and DU 145, and was highest in LNCaP. In cell culture, cdk4 was lowest in Calu-6, HCT116, HT29 and DU-145 and highest in H82 and SW2; in tumors, cdk4 was low in MCF-7, MB-468, H460, Calu-6 and HCT116 and was very high in the SW2. Expression of cyclin E was very low in MCF-7 and HT29 and high in H460 in culture and was very low in MCF-7, H460, Calu-6, H82, HT29 and DU-145 in tumors and high in HCT116 and LNCaP. In cell culture, E2F1 was lowest in MB-468, Calu-6, HT29 and DU-145 cells and highest in LNCaP cells; in tumors, E2F1 was lowest in MCF-7, MB-468 and Calu-6 and highest in LNCaP. In cell culture, DP-1 was lowest in MB-468, HCT116 and HT29 and highest in SW2. The MCF-7 and MB-468 lines were most resistant to flavopiridol and olmoucine and the H460 and Calu-6 lines were most resistant to genistein. The SW2 tumor was most responsive to flavopiridol and olomoucine. CONCLUSIONS: There is a high degree of variability in the expression of cell cycle components in human tumor cell lines, resulting in complexity in predicting response to cell cycle directed agents.


Subject(s)
Calmodulin-Binding Proteins/biosynthesis , Cell Cycle Proteins , Cyclin-Dependent Kinases/biosynthesis , Cyclins/biosynthesis , Plant Proteins/biosynthesis , Transcription Factors/biosynthesis , Aged , Animals , Antineoplastic Agents/pharmacology , Blotting, Western , Cell Cycle/drug effects , Cell Survival/drug effects , Cyclin-Dependent Kinases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Female , Flavonoids/pharmacology , Genistein/pharmacology , Humans , Kinetin , Male , Mice , Mice, Nude , Middle Aged , Neoplasm Transplantation , Piperidines/pharmacology , Purines/pharmacology , Transcription Factor DP1 , Transplantation, Heterologous , Tumor Cells, Cultured
13.
Cancer Chemother Pharmacol ; 46(2): 142-9, 2000.
Article in English | MEDLINE | ID: mdl-10972484

ABSTRACT

INTRODUCTION/PURPOSE: Cryptophycins are a family of antitubulin antitumor agents. A synthetic cryptophycin derivative (LY355703, CRYPTO 52) is in early clinical evaluation. The effect of infusion time on the antitumor activity of four cryptophycins was assessed in rats bearing the 13762 mammary carcinoma and combination treatment regimens were assessed in nude mice bearing human tumor xenografts. METHODS: The cryptophycins were prepared in 2% PEG300/8% cremophor/90% normal saline and delivered by jugular vein catheter on days 7, 9 and 11 post tumor implant to 13762 tumor-bearing rats. The cryptophycins prepared in the same formulation were administered by intravenous bolus injection on an alternate day schedule for five doses to human tumor xenograft bearing nude mice. RESULTS: An infusion time of 2 h in the rats increased the tumor growth delay produced by CRYPTO 52 and CRYPTO 55, while increasing the infusion time to 6 h continued to increase the tumor growth delay for CRYPTO 292 and CRYPTO 296. Administering CRYPTO 292 at a higher dose two times was more effective than administering it at a lower dose three times. The tumor growth delays produced by the cryptophycins in the rat 13762 mammary carcinoma were greater than those with cisplatin, doxorubicin, 5-fluorouracil and 5 x 3 Gray and comparable with cyclophosphamide and gemcitabine. Combination studies were carried out in human tumor xenografts including the MX-1 breast carcinoma, the Calu-6 non-small cell lung carcinoma, the H82 small cell lung carcinoma and the SW-2 small cell lung carcinoma. CRYPTO 52 and CRYPTO 55 combined with doxorubicin, paclitaxel and 5-fluorouracil to form highly effective regimens against the human MX-1 breast carcinoma. CRYPTO 52 and CRYPTO 55 were also highly effective against the three lung carcinoma xenografts when combined with the antitumor platinum complexes, cisplatin, carboplatin or oxaliplatin. CONCLUSIONS: Cryptophycins represent a promising new class of antitumor agents that may be optimally administered by intravenous infusion and in combination with doxorubicin, paclitaxel and 5-fluorouracil.


Subject(s)
Antineoplastic Agents/therapeutic use , Depsipeptides , Lactams/therapeutic use , Lactones/therapeutic use , Lung Neoplasms/drug therapy , Mammary Neoplasms, Experimental/drug therapy , Animals , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma/drug therapy , Carcinoma/pathology , Female , Humans , Infusions, Intravenous , Lactams/administration & dosage , Lactones/administration & dosage , Lung Neoplasms/pathology , Male , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Nude , Peptides, Cyclic/administration & dosage , Peptides, Cyclic/therapeutic use , Rats , Rats, Inbred F344 , Transplantation, Heterologous
14.
In Vivo ; 14(4): 471-80, 2000.
Article in English | MEDLINE | ID: mdl-10945160

ABSTRACT

The antitumor activity of cryptophycin 52 (C52) and cryptophycin 55 (C55) in sequential and simultaneous combination treatment regimens in human tumor xenografts models was explored. The antitumor activity of C52 and C55 was compared alone and in sequential combination with gemcitabine or paclitaxel in four lung cancer models, H460 and Calu-6 NSCLC and SW2 and H82 small cell lung carcinoma. The combination of C52 followed by gemcitabine was additive in three tumors and greater-than-additive in the fourth. The combination of C55 followed by gemcitabine was additive in three tumors and less-than-additive in the fourth. The combination of C52 followed by paclitaxel was greater-than-additive in one tumor, additive in one tumor and less-than-additive in two tumors. The combination of C55 followed by paclitaxel was greater-than-additive in two tumors and less-than-additive in two tumors. The simultaneous combination of C52 or C55 with fractionated radiation therapy was assessed in the H460 NSCLC tumor. Both cryptophycins produced a tumor response that was additive along with radiation therapy. The HCT116 colon carcinoma was used to compare the antitumor activity of simultaneous or sequential combination of 5-fluorouracil or irinotecan with C52. C52 produced greater-than-additive tumor response when administered either simultaneously with or sequentially with 5-fluorouracil or iriniotecan. Finally, when administered to animals bearing intraperitoneal OVCAR-3 ovarian carcinoma, C52, docetaxel and paclitaxel resulted in mean survival times of 123, 80 and 85 days compared with 72 days in the untreated controls. In combination with carboplatin, C52, docetaxel and paclitaxel resulted in mean survival times of 140, 105 and 135 days. Cryptophycins have the potential to be useful chemotherapeutic agents in a wide variety of clinical combinations regimens.


Subject(s)
Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Depsipeptides , Lactams/therapeutic use , Lactones/therapeutic use , Lung Neoplasms/drug therapy , Peptides, Cyclic/therapeutic use , Animals , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/therapeutic use , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/therapeutic use , Antineoplastic Agents/administration & dosage , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Female , Humans , Lactams/administration & dosage , Lactones/administration & dosage , Lung Neoplasms/pathology , Male , Mice , Mice, Nude , Paclitaxel/administration & dosage , Paclitaxel/therapeutic use , Peptides, Cyclic/administration & dosage , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Gemcitabine
15.
Clin Cancer Res ; 6(3): 1016-23, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10741729

ABSTRACT

The scheduling of antifolate antitumor agents, including the new multitargeted autofolate LY231514 (MTA), with 5-fluorouracil was explored in the human MX-1 breast carcinoma and human H460 and Calu-6 non-small cell lung carcinoma xenografts to assess antitumor activity and toxicity (body weight loss). Administration of the antifolate (methotrexate, MTA, or LY309887) 6 h prior to administration of 5-fluorouracil resulted in additive growth delay of the MX-1 tumor when the antifolate was methotrexate or LY309887 and greater-than-additive tumor growth delay (TGD) when the antifolate was MTA. In the H460 tumor, the most effective regimens were a 14-day course of MTA or LY309887 along with 5-fluorouracil administered on the final 5 days. In addition, the simultaneous combination of MTA administered daily for 5 days for 2 weeks with administration of gemcitabine resulted in greater-than-additive H460 TGD. MTA was additive with fractionated radiation therapy in the H460 tumor when the drug was administered prior to each radiation fraction. MTA administered along with paclitaxel produced greater-than-additive H460 TGD and additive responses along with vinorelbine and carboplatin. In the Calu-6 non-small cell lung carcinoma xenograft, MTA administered in combination with cisplatin or oxaliplatin was highly effective, whereas MTA administered in combination with cyclophosphamide, gemcitabine, or doxorubicin produced additive responses. Administration of MTA along with paclitaxel or doxorubicin resulted in additive MX-1 TGD. Thus, MTA appears to be especially effective in combination therapies including 5-fluorouracil or an antitumor platinum complex.


Subject(s)
Breast Neoplasms/prevention & control , Carcinoma, Non-Small-Cell Lung/prevention & control , Folic Acid Antagonists/therapeutic use , Glutamates/therapeutic use , Guanine/analogs & derivatives , Lung Neoplasms/prevention & control , Taxoids , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/pathology , Carboplatin/administration & dosage , Carcinoma, Non-Small-Cell Lung/pathology , Cell Division/drug effects , Cisplatin/administration & dosage , Cyclophosphamide/administration & dosage , Docetaxel , Female , Fluorouracil/administration & dosage , Glutamates/administration & dosage , Guanine/administration & dosage , Guanine/therapeutic use , Humans , Injections, Intraperitoneal , Lung Neoplasms/pathology , Male , Mice , Mice, Nude , Neoplasm Transplantation , Organoplatinum Compounds/administration & dosage , Oxaliplatin , Paclitaxel/administration & dosage , Paclitaxel/analogs & derivatives , Pemetrexed , Transplantation, Heterologous , Tumor Cells, Cultured , Vinblastine/administration & dosage , Vinblastine/analogs & derivatives , Vinorelbine , Weight Loss/drug effects
16.
Int J Oncol ; 16(3): 599-610, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10675495

ABSTRACT

Liver cancer and gastric cancer are the most common solid tumors worldwide. Transforming growth factor-beta (TGF-beta) production and lack of response to TGF-beta growth inhibitory effects have been associated with tumor progression and therapeutic resistance. HepG2, Hep3B, and SK-HEP-1 human liver cancer lines produce 3, 5.7, and 2.5 ng TGF-beta1; 1.4, 2, and 4 ng TGF-beta2 and 0.15, 0.2 and 0.22 ng TGF-beta3 per 107 cells (24 h). Expression of the TGF-beta type I receptor is 20x, 1x, and 0.6x the level in mink lung MvLu1 cells in the HepG2, Hep3B, and SK-HEP-1 cells, respectively. HepG2 and Hep3B cells do not express the TGF-beta type II receptor while SK-HEP-1 cells express 7x the level found in mink lung MvLu1 cells. Hs 746T, KATO III, RF-1, and RF-48 human gastric cancer cell lines produce 12. 5, 0.35, 0.4, and 0.4 ng TGF-beta1; 2.6, 0.95, 0.5, and 0.52 ng TGF-beta2 and 0.42, 0.17, 0.12, and 0.14 ng TGF-beta3 per 107 cells (24 h). Expression of TGF-beta type I receptor is 0.7x, 0.7x, 0.8x, 0.6x the level in mink lung MvLu1 cells in the Hs 746T, KATO III, RF-1 and RF-48 cells, respectively. KATO III cells are lacking in the TGF-beta type II receptor while Hs 746T, RF-1 and RF-48 cells express 10x, 0.8x, and 1x the levels in mink lung MvLu1 cells. The IC50 for TGF-beta1 is >>10 ng/ml in all of these lines except RF-48 where TGF-beta1 is mitogenic. The response of the cell lines to radiation, doxorubicin, mitomycin C, cisplatin, 5-fluorouracil, methotrexate, and gemcitabine showed that SK-HEP-1 was the most drug resistant liver cancer cell line and KATO III was the most drug resistant gastric cancer cell line. Overall, there was no correlation between TGF-beta secretion in cell culture and sensitivity of the cells to anticancer agents. Increased TGF-beta1 levels were detectable in the plasma of nude mice bearing Hep3B and Hs 746T xenografts. Those tumors which secreted greater amounts of TGF-beta were more therapeutically resistant in vivo.


Subject(s)
Activin Receptors, Type I , Antineoplastic Agents/therapeutic use , Liver Neoplasms/drug therapy , Stomach Neoplasms/drug therapy , Transforming Growth Factor beta/physiology , Adolescent , Aged , Animals , Child , Humans , Liver Neoplasms/pathology , Male , Mice , Middle Aged , Protein Serine-Threonine Kinases/analysis , Receptor, Transforming Growth Factor-beta Type I , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/analysis , Stomach Neoplasms/pathology , Transforming Growth Factor beta/pharmacology , Tumor Cells, Cultured
17.
Semin Oncol ; 26(2 Suppl 6): 55-62, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10598556

ABSTRACT

An important component in the development of a new anticancer drug is an understanding of its potential for inclusion in combination treatment regimens. LY231514, a multitargeted antifolate (MTA), was tested in combination with cisplatin, methotrexate, 5-fluorouracil, paclitaxel, docetaxel, doxorubicin, LY329201 (a glycinamide ribonucleotide formyltransferase [GARFT] inhibitor), and fractionated radiation therapy in vivo using EMT-6 mammary carcinoma, human HCT 116 colon carcinoma, and human H460 nonsmall cell lung carcinoma grown as xenografts in nude mice. Isobologram methodology was used to determine the additivity or synergy of the combination regimens. MTA administered with cisplatin, paclitaxel, docetaxel, or fractionated radiation therapy produced additive to greater than additive tumor response by tumor cell survival assay and tumor growth delay. While an additive tumor response was observed when MTA was administered with methotrexate, synergistic tumor responses were seen when MTA was administered with the GARFT inhibitor, LY329201, or with the topoisomerase I inhibitor, irinotecan. MTA was administered in combination with full doses of each anticancer agent studied, with no evidence of increased toxicity resulting from the combination.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Enzyme Inhibitors/therapeutic use , Folic Acid Antagonists/therapeutic use , Glutamates/administration & dosage , Guanine/analogs & derivatives , Neoplasms, Experimental/drug therapy , Thymidylate Synthase/antagonists & inhibitors , Animals , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/radiotherapy , Colonic Neoplasms/drug therapy , Colonic Neoplasms/radiotherapy , Combined Modality Therapy , Glutamates/therapeutic use , Guanine/administration & dosage , Guanine/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/radiotherapy , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/radiotherapy , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Neoplasms, Experimental/radiotherapy , Pemetrexed , Tumor Cells, Cultured
18.
Cancer Res ; 59(22): 5836-41, 1999 Nov 15.
Article in English | MEDLINE | ID: mdl-10582707

ABSTRACT

Using very low frequency in vivo electron paramagnetic resonance (EPR), we have compared, for the first time, the average microviscosity of the total aqueous compartment of murine fibrosarcomas and that of normal leg tissue in a living animal. EPR spectra from dissolved nitroxide spin probes report the solvent microviscosity. The tumor aqueous microviscosity, 1.8 +/- 0.1 centipoise, was significantly lower than that of the corresponding normal tissue, 2.9 +/- 0.3 centipoise, a difference of 38 +/- 7%. These results confirm the commonly observed increase in the water proton transverse relaxation times (T2) in magnetic resonance imaging of hyperproliferative states, for example, malignancy. The specificity of the localization of the EPR signal indicates a substantial portion of the T2 increase seen in magnetic resonance imaging derives from decreased bulk-water viscosity. The effect of this microviscosity differences may be the basis of several physiological differences between tumors and normal tissues which could confer a growth rate advantage to tumor tissue.


Subject(s)
Body Water/chemistry , Neoplasms, Experimental/chemistry , Animals , Electron Spin Resonance Spectroscopy , Mice , Mice, Inbred C3H , Neoplasms, Experimental/physiopathology , Viscosity
19.
Adv Enzyme Regul ; 39: 313-27, 1999.
Article in English | MEDLINE | ID: mdl-10470381

ABSTRACT

The macrocyclic bisindolylmaleimide, LY333531, selectively inhibits protein kinase C beta 1 and beta 2 isoforms with an approximate IC50 of 5 nanomolar. The efficacy of LY333531 administered alone and in combination with cytotoxic cancer therapies in models of non-small cell lung carcinoma and brain tumors was determined in vivo. In the Lewis lung carcinoma, administration of LY333531 enhanced the activity of paclitaxel and fractionated radiation and, to a lesser degree, carboplatin and gemcitabine. In the human T98G glioblastoma multiforme xenograft, the addition of LY333531 to treatment with carmustine (BCNU) resulted in enhanced tumor response in a nodule grown subcutaneously and increased life-span in animals bearing an intracranial tumor from 37 days in the control animals to 64 days in the BCNU treated animals, and to 104 days in the LY333531 plus BCNU treated animals with 4 out of 5 animals being long-term survivors.


Subject(s)
Enzyme Inhibitors/therapeutic use , Isoenzymes/antagonists & inhibitors , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/enzymology , Protein Kinase C/antagonists & inhibitors , Animals , Antineoplastic Agents/administration & dosage , Brain Neoplasms/drug therapy , Carcinoma, Non-Small-Cell Lung/drug therapy , Combined Modality Therapy , Drug Therapy, Combination , Enzyme Inhibitors/administration & dosage , Female , Glioblastoma/drug therapy , Humans , Indoles/administration & dosage , Indoles/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Male , Maleimides/administration & dosage , Maleimides/therapeutic use , Mice , Mice, Inbred C57BL , Mice, Nude , Neoplasm Transplantation , Neoplasms, Experimental/radiotherapy , Protein Kinase C beta , Transplantation, Heterologous
20.
Clin Cancer Res ; 5(9): 2638-45, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10499643

ABSTRACT

The anticancer activity of the boronic acid dipeptide proteasome inhibitor PS-341 was examined in vitro and in vivo. PS-341 was a potent cytotoxic agent toward MCF-7 human breast carcinoma cells in culture, producing an IC90 of 0.05 microM on 24 h of exposure to the drug. In the EMT-6 tumor cell survival assay, PS-341 was equally cytotoxic administered p.o. or by i.p. injection up to a dose of 2 mg/kg. PS-341 was also toxic to the bone marrow colony-forming unit-granulocyte macrophage. PS-341 increased the tumor cell killing of radiation therapy, cyclophosphamide, and cisplatin in the EMT-6/Parent tumor, but was not able to overcome the in vivo resistance of the EMT-6/CTX and EMT-6/CDDP tumors. In the tumor growth delay assay, PS-341 administered p.o. had antitumor activity against the Lewis lung carcinoma, both primary and metastatic disease. In combination, regimens with 5-fluorouracil, cisplatin, Taxol and adriamycin, PS-341 seemed to produce primarily additive tumor growth delays against the s.c. tumor and was highly effective against disease metastatic to the lungs. The proteasome is an interesting new target for cancer therapy, and the proteasome inhibitor PS-341 warrants continued investigation in cancer therapy.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/pharmacology , Boronic Acids/pharmacology , Breast Neoplasms/drug therapy , Dipeptides/pharmacology , Protease Inhibitors/pharmacology , Pyrazines/pharmacology , Adenocarcinoma/radiotherapy , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Boronic Acids/metabolism , Bortezomib , Breast Neoplasms/radiotherapy , Cisplatin/administration & dosage , Cyclophosphamide/administration & dosage , Drug Synergism , Humans , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/radiotherapy , Mice , Mice, Inbred BALB C , Radiation-Sensitizing Agents/pharmacology , Tumor Cells, Cultured , Ubiquitins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...