Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Anticancer Res ; 28(5A): 2769-74, 2008.
Article in English | MEDLINE | ID: mdl-19035308

ABSTRACT

BACKGROUND: The somatostatin structural derivative, TT-232, has a special 5-residue ring structure (D-Phe-Cys-Tyr-D-Trp-Lys-Cys-Thr-NH2) and very different characteristics from the known growth hormone (GH) active somatostatin analogs. This somatostatin structural derivative has no GH release inhibitory or antisecretory activity and does not bind to rat pituitary or the cortex, where all the known somatostatin receptor subtypes are expressed. TT-232 had previously been shown to inhibit the proliferation of a large number of cancer cell lines in vitro and reduce the size of different tumors in animal models in vivo. MATERIALS AND METHODS: The therapeutic efficacy of TT-232 was evaluated in different long-term administration routes: the traditional injection (i.p. or s.c.) versus infusion treatment via s.c.- or i.v.-inserted Alzet osmotic minipump, and on different types of transplantable rodent (S-180 sarcoma, P-388sc lymphoid leukemia, Colon-26 adenocarcinoma, MXT breast carcinoma, B-16 melanoma) and human tumor models (HT-18 lymphoid melanoma, T-47/D breast carcinoma, A-431 epidermoid carcinoma). On the basis of our previous experiments the optimum injected dose of TT-232 was found to be 15 microg/kg twice a day. This dose is equivalent to 0.6 dg/day by infusion therapy. RESULTS: In our experiments, the best results were achieved when TT-232 was applied as an infused treatment. In the S-180 sarcoma and P-388sc lymphoid leukemia rodent tumor models the infusion treatment with TT-232 resulted in 61%-100% tumor growth inhibition and in 20%-60% of the mice being long-term and tumor-free survivors. In the aggressive Colon-26 adenocarcinoma and MXT breast carcinoma models, the infusion treatment resulted in 52%-75% tumor growth inhibition. In the B-16 melanoma model, the infusion treatments resulted in 47% -63% growth inhibition. The tumor growth inhibitory effect of infusion treatment with TT-232 on HT-18 human lymphoid melanoma tumor proved to be significant, resulting in 69%-79% decreases in tumor volume. In the T-47/D human breast carcinoma, the infusion treatment resulted in 48%-53% tumor growth inhibition. The tumor growth inhibitory effect of infusion treatment on A-431 human epidermoid carcinoma tumor resulted in 70%-74% decreases in tumor volume. CONCLUSION: The antitumor efficacy of TT-232 was seen in almost all the tumors investigated. In our study, the route of infusion was shown to increase drug efficacy relative to conventional delivery methods (injection). The results obtained from this study suggest that TT-232 is a promising new antitumor agent in cancer chemotherapy and a good candidate for delivery by continuous (infusion) therapy.


Subject(s)
Neoplasms, Experimental/drug therapy , Peptides, Cyclic/administration & dosage , Animals , Humans , Infusion Pumps, Implantable , Infusions, Subcutaneous , Mice , Somatostatin/analogs & derivatives
2.
Anticancer Res ; 27(6B): 4015-9, 2007.
Article in English | MEDLINE | ID: mdl-18225564

ABSTRACT

BACKGROUND: The somatostatin structural deivative, TT-232, has a special 5'-residue ring structure (D-Phe-Cys-Tyr-D-Trp-Lys-Cys-Thr-NH2) and very different characteristics from the known growth hormone (GH) active somatostatin analogs. TT-232 inhibited tyrosine kinase activity of tumor cell lines and this inhibition correlated well with the inhibition of cell proliferation of a large number of cancer cell lines in vitro and reduces the size of different tumors in animal models in vivo. The antitumor efficacy of TT-232 has been found to be associated with the induction of apoptosis in tumor cells, resulting in highly selective elimination of tumor tissue. TT-232 was found to be devoid of GH release inhibitory activity but to possess strong antitumor effects. It binds with a high affinity to SSTR1 and SSTR4. This compound was also found to inhibit inflammation in a number of experimental models. MATERIALS AND METHODS: The study compared the antitumor effect of TT-232 in various long-term administration routes: an intermittent (injection) versus continuous (infusion) treatment via subcutaneously inserted 2002 type Alzet osmotic minipumps in two different tumor models (B-16 rodent melanoma and HT-18 human lymphoid melanoma). Treatment with TT-232 started after disease development. The antitumor efficacy of TT-232 was evaluated on the basis of tumor growth inhibition and survival time. RESULTS: In the case of B-16 rodent melanoma, the TT-232 treatments resulted in 35%-39% (injection) and 47%-63% (infusion) tumor growth inhibition, and the infusion treatment an approximately 61% increase in survival time. The tumor growth inhibitory effect of TT-232 on HT-18 lymphoid melanoma tumor proved to be significant, resulting in 41%-63% (injection) and 69%-79% (infusion) decreases in tumor volume and in a 25%-30% increase in survival time (infusion treatments). CONCLUSION: The results indicate that TT-232 could be a potentially useful therapeutic agent if these data are translated into clinical practice.


Subject(s)
Melanoma, Experimental/drug therapy , Melanoma/drug therapy , Peptides, Cyclic/pharmacology , Somatostatin/analogs & derivatives , Animals , Antineoplastic Agents/pharmacology , Drug Administration Routes , Humans , Infusion Pumps , Mice
3.
Anticancer Res ; 26(5A): 3477-83, 2006.
Article in English | MEDLINE | ID: mdl-17094470

ABSTRACT

The antitumor effects of the somatostatin structural derivative TT-232 in different rodent and xenograft tumor models are summarized in this report. TT-232 had previously been shown to inhibit the proliferation of a large number of cancer cell lines in vitro and reduce the size of different tumors in animal models in vivo. The effects of TT-232 by different routes of administration and treatment schedules were studied in various types of rodent and human xenograft tumor models. In the rodent tumor models S-180 sarcoma and P-388 lymphoid leukemia tumor the infusion treatment resulted in 76%-100% tumor growth inhibition and in 20%-60% of the mice being long-term and tumor-free survivors. In the aggressive C-26 colon carcinoma and MXT breast carcinoma, the TT-232 treatments resulted in 71%-75% tumor growth inhibition and an approximately 50% increased survival time. The tumor growth inhibitory effect of TT-232 on human tumor xenografts proved to be significant, resulting in 30%-80% decrease in tumor volume and in 20%-40% tumor-free animals. This antitumor efficacy of TT-232 was seen in almost all the tumors investigated. In our study, the route of infusion was shown to increase drug efficacy relative to conventional delivery methods. Our results suggested that TT-232 is an effective and promising antitumor agent.


Subject(s)
Colonic Neoplasms/drug therapy , Leukemia P388/drug therapy , Mammary Neoplasms, Experimental/drug therapy , Peptides, Cyclic/therapeutic use , Sarcoma 180/drug therapy , Animals , Colonic Neoplasms/pathology , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Screening Assays, Antitumor , HL-60 Cells , HT29 Cells , Humans , Leukemia P388/pathology , Male , Mammary Neoplasms, Experimental/pathology , Melanoma/drug therapy , Mice , Mice, Inbred Strains , Prostatic Neoplasms/drug therapy , Sarcoma 180/pathology , Somatostatin/analogs & derivatives , Transplantation, Heterologous , Tumor Cells, Cultured
4.
Anticancer Res ; 26(4B): 3011-5, 2006.
Article in English | MEDLINE | ID: mdl-16886628

ABSTRACT

The tumor growth inhibitory efficacy of the somatostatin structural derivative TT-232 was studied using different routes of administration and treatment schedules in various human tumor models. TT-232, containing a five-residue ring structure, has a strong antitumor activity both in vitro and in vivo. The antineoplastic activity of TT-232 has been found to be associated with the induction of programmed cell death in tumor cells, resulting in highly-selective elimination of the neoplastic tissue. The study compared the antitumor efficacy of TT-232 in various long-term administration routes; the intermittent (injection) versus continuous (infusion) treatment via subcutaneously-inserted Alzet osmotic minipumps in different human tumor models: T-47/D human breast carcinoma and A-431 human epidermoid carcinoma. Treatment with TT-232 started after disease development. The antitumor activity of TT-232 was evaluated on the basis of the tumor growth inhibition. In the case of T-47/D human breast carcinoma, the intermittent treatment resulted in 23%-26% and the infusion treatment resulted in 48%-53% tumor growth inhibition. The tumor growth inhibitory effect of TT-232 on A-431 human epidermoid carcinoma tumor resulted in 35%-43% (intermittent treatment) and 70%-74% (continuous treatment) decreases in tumor volume. This antitumor efficacy of TT-232 was observed in the two human tumors investigated. In this study, the route of infusion was shown to increase drug efficacy relative to conventional delivery methods. The results suggest that TT-232 is an effective and promising antitumor agent.


Subject(s)
Breast Neoplasms/drug therapy , Carcinoma, Squamous Cell/drug therapy , Peptides, Cyclic/administration & dosage , Animals , Antineoplastic Agents/administration & dosage , Cell Growth Processes/drug effects , Drug Administration Schedule , Drug Screening Assays, Antitumor , Female , Humans , Mice , Mice, SCID , Somatostatin/analogs & derivatives , Xenograft Model Antitumor Assays
5.
Anticancer Res ; 25(1A): 325-30, 2005.
Article in English | MEDLINE | ID: mdl-15816555

ABSTRACT

TT-232 is a structural derivative of the natural signal inhibitory peptide somatostatin, with selective antiproliferative and anti-inflammatory properties. TT-232 activates SSTR receptors (primarily the SSTR-1), which leads to irreversible cell cycle arrest, followed by secondary induction of apoptosis. TT-232 has passed phase I clinical trials without toxicity and significant side-effects. We examined the antiproliferative effect in vitro and the antitumor effect in vivo of TT-232 on leukemia cell lines. During in vivo experiments, we evaluated the therapeutic efficacy of TT-232 in various long-term administration routes; traditional injection versus infusion treatment via an inserted Alzet minipump on P-388 mice and HL-60 human leukemia models. Treatment with TT-232 started after development of the disease. In vitro, TT-232 inhibited the proliferation of P-388 mice lymphoid cells and HL-60 human promyelocytic leukemia cells in the range of 46%-97% with 24-hour treatment and 82%-100% with 48-hour treatment. Cells were treated with 30 microg/ml and 60 microg/ml dose of TT-232. With the same in vivo models, the best results were achieved when TT-232 was applied by infusion treatments. The infusion treatment with TT-232 produced 50%-80% inhibition of growth and resulted in 20%-40% long-term and leukemia-free survivors. TT-232 showed dose-, time- and administration mode-dependent antileukemia activity in vitro and in vivo, both on rodent and human models. Our results suggest that TT-232 is a promising new antileukemia agent.


Subject(s)
Leukemia P388/drug therapy , Leukemia, Promyelocytic, Acute/drug therapy , Peptides, Cyclic/pharmacology , Animals , Antineoplastic Agents/pharmacology , Female , Growth Inhibitors/pharmacology , HL-60 Cells , Humans , Mice , Mice, Inbred CBA , Somatostatin/analogs & derivatives
6.
Anticancer Res ; 24(2B): 865-71, 2004.
Article in English | MEDLINE | ID: mdl-15161039

ABSTRACT

The efficiency of chemotherapy is often decreased by the development of resistance of cancer cells to cytostatic drugs. This phenomenon is in most cases caused by the activity of the various ABC transporters, multidrug-resistance (MDR) gene-encoded p-glycoproteins, that pump anticancer drugs out of the cells. The inhibition of the activities of the MDR proteins MDR1 and MRP was investigated via the administration of two new organosilicon compounds, alis-409 and alis-421. The study was focused on the inhibition of MDR by blocking the ADR1 gene expression and through the inhibition of the pump-function of mdr-p-glycoprotein, in human breast cancer cell lines expressing mrp and prostate cancer cell line (PC-3). Apoptosis induction and the interaction between epirubicin and the silicon-substituted compounds were studied in human MDR-1 gene-transfected mouse lymphoma and its parent cell line, Colo320/MDR-LRP and sensitive subline Colo205, by means of rhodamine 123 accumulation. The activity of MRP1 p-glycoprotein was studied in human breast cancer cell lines such as HTB-26/MRP1 and two MRP-negative breast cancer cell lines, T47D and MCF7, by carboxyfluorescein accumulation, and on a stomach cancer cell line. The activity of MRP in 257P/MDR and its drug-sensitive derivative were studied in human stomach cancer cells by daunorubicin accumulation in a flow cytometer. The two representative organosilicon derivatives, alis-409 and alis-421, showed antiproliferative effects without apoptosis induction. The drug accumulation in the human MDR1 gene-transfected mouse lymphoma cells was increased without down-regulation of the MDR1 gene expression tested by RT-PCR assay. The rhodamine uptake was increased in L5178/MDR1 and Colo320/MDR1-LRP, but not drug-sensitive human breast cancer MCF-7 and T47D, and L5178 mouse lymphoma parent cells in the presence of alis-409 and alis-421. The MRP-mediated carboxyfluorescein accumulation in HTB-26/MRP human breast cancer cells and daunorubicin accumulation in human stomach cancer cells 257P/MDR were not modified by these alis compounds. A synergistic interaction between epirubicin and the silicon-substituted resistance modifiers was found only in MDR1-mediated MDR in the case of colo-320/MDR1-LRP cells and mouse lymphoma cells transfected with the human MDR1 gene. The results indicate that the organosilyl derivatives specifically act on MDR1 p-glycoprotein 170. The alis compounds act on pgp170 in a way which is similar to verapamil isomers.


Subject(s)
Drug Resistance, Multiple/drug effects , Neoplasms/drug therapy , Organosilicon Compounds/pharmacology , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Division/drug effects , Cell Line, Tumor , Drug Resistance, Multiple/genetics , Epirubicin/pharmacology , Fluoresceins/pharmacokinetics , Genes, MDR/genetics , Humans , K562 Cells , Lymphoma, T-Cell/drug therapy , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/pathology , Male , Mice , Neoplasms/genetics , Neoplasms/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Rhodamine 123/pharmacokinetics , Transfection
7.
Anticancer Res ; 23(5A): 4061-6, 2003.
Article in English | MEDLINE | ID: mdl-14666719

ABSTRACT

A somatostatin structural derivative (TT-232) has been developed in our laboratory with strong antiproliferative effect but no GH- release inhibitory activity. TT-232 inhibited tyrosine kinase activity of tumor cells lines and this inhibition correlated well with the inhibition of cell proliferation. The antineoplastic activity of TT-232 has been found to be associated with induction of programmed cell death (apoptosis) in tumor cell, resulting in highly selective elimination of neoplastic tissue. The aim of this study was the therapeutic efficacy of TT-232 on different human models: PC-3 prostate carcinoma, MDA-MB-231 (ER-) and MCF-7 (ER+) breast carcinoma, HT-29 colon carcinoma, HT-18 melanoma, HL-60 promyelocytic leukemia. We studied the therapeutic efficacy of the novel somatostatin analog, it for 30 days with intermittent injection once daily and for 14 days with s.c. infusion using the Alzet osmotic minipump (model 2002). The antitumor activity of TT-232 was evaluated on the basis of survival time and tumor growth inhibition. The tumor growth inhibitory effect of TT-232 on human tumor xenografts proved to be significant, resulting in 30%-80% decrease in tumor volume and in 20-60% tumor free animals. This antitumor efficacy of the novel somatostatin analog was observable in almost all tumors investigated. These data suggest that the novel somatostatin analog (TT-232) is an effective and promising antitumor agent.


Subject(s)
Antineoplastic Agents/pharmacology , Peptides, Cyclic/pharmacology , Breast Neoplasms/drug therapy , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Drug Screening Assays, Antitumor , HL-60 Cells , HT29 Cells , Humans , Male , Melanoma/drug therapy , Prostatic Neoplasms/drug therapy , Somatostatin/analogs & derivatives , Xenograft Model Antitumor Assays
8.
Redox Rep ; 6(2): 99-103, 2001.
Article in English | MEDLINE | ID: mdl-11450989

ABSTRACT

Telomerase is a specialized ribonucleoprotein enzyme complex which prevents the loss of the telomere. The activity of telomerase can be up- and down-regulated by various oxidative stresses but the effect of physical exercise is not known, whereas the modifying effect of cancer on telomerase activity is well documented. In the first study, we investigated the effect of mild and strenuous exercise training on telomerase activity, assessed by a PCR ELISA kit. No alteration in telomerase activity was detected. In the second investigation, solid sarcoma cells were transplanted to control, exercise trained or exercise trained and still exercising mice. On the 16th day after the transplantation, the size of tumors in the exercise trained group was 72% and in the exercising group 57% (P < 0.05) of that in the controls. Telomerase activity and 8-hydroxy-2'-deoxyguanosine levels in the liver were not significantly altered by exercise and/or sarcoma. We conclude that mild and strenuous exercise training does not significantly affect the activity of telomerase in the systems studied. Exercise training during sarcoma significantly retards the development of tumors and could possibly serve as a positive adjunct to treatment.


Subject(s)
Deoxyguanosine/analogs & derivatives , Liver Neoplasms, Experimental/enzymology , Liver/enzymology , Muscle, Skeletal/enzymology , Physical Conditioning, Animal/physiology , Physical Exertion/physiology , Sarcoma, Experimental/enzymology , Stress, Physiological/enzymology , Telomerase/metabolism , 8-Hydroxy-2'-Deoxyguanosine , Animals , Crosses, Genetic , DNA Damage , Deoxyguanosine/analysis , Female , Liver Neoplasms, Experimental/chemistry , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Neoplasm Transplantation , Oxidative Stress , Rats , Rats, Wistar , Sarcoma, Experimental/chemistry
9.
Anticancer Res ; 20(2A): 1023-7, 2000.
Article in English | MEDLINE | ID: mdl-10810391

ABSTRACT

TT-232 a novel tumor-selective somatostatin analog with a five residue ring structure (D-Phe-Cys-Tyr-D-Trp-Lys-Cys-Thr-NH2) was developed by us and published in an earlier work. This synthetic heptapeptide had no effect on growth hormone release, but had a remarkable tyrosine-kinase inhibitory effect and inducted apoptosis. The aim of this study was to compare the therapeutic efficacy of TT-232 used in various long-term administration routes and treatment schedules. The effectiveness of TT-232 was studied on different rodent tumors transplanted to inbred mice from SPF breeding. Intermittent treatment by injections and continuous infusion of TT-232 using a s.c., i.p. or i.v. implanted Alzet type osmotic minipump were compared for therapeutic efficacy. The treatments were started either on the day subsequent to tumor transplantation or after the development of a tumor. On the basis of survival and tumor growth inhibition the infusion of TT-232 for 14 days using an implantable osmotic pump proved to be a much more effective route of treatment in both s.c. and i.v. administration than the intermittent injections applied twice a day for 2 weeks. In the case of S-180 sarcoma the continuous administration of TT-232 for 14 days using s.c. implanted osmotic pump resulted in 60% the i.v. infusion produced 40% long-term (over 80 days) and tumor free survivors. By the continuous administration of TT-232, an 80-100% tumor growth inhibitory effect and a considerable retardation of tumor development could be achieved. Continuous infusion from implanted pumps ensured a constant drug level and resulted in a well-defined, consistent pattern of drug exposure over the full duration of drug administration. In our study the route of infusion has been shown to increase drug efficacy relative to conventional delivery methods.


Subject(s)
Antineoplastic Agents/administration & dosage , Peptides, Cyclic/administration & dosage , Sarcoma 180/drug therapy , Animals , Antineoplastic Agents/therapeutic use , Female , Infusions, Parenteral , Injections, Intravenous , Injections, Subcutaneous , Mice , Mice, Inbred Strains , Peptides, Cyclic/therapeutic use , Sarcoma 180/pathology , Somatostatin/analogs & derivatives
10.
J Control Release ; 63(1-2): 81-95, 2000 Jan 03.
Article in English | MEDLINE | ID: mdl-10640582

ABSTRACT

In a new group of polypeptides, the branches were composed of DL-Ala oligopeptide, L-serine and L-leucine or L-glutamic acid residues. The synthesis of eight different side-chain combinations is described. In the first group, Ser was attached directly to the epsilon-amino groups of polylysine, and Leu or Glu was situated at the side chain end (poly[Lys(X(i)-DL-Ala(m)-Ser(j))]). Alternatively, Leu or Glu was positioned next to the polylysine backbone (poly[Lys(Ser(j)-DL-Ala(m)-X(i))], where X=L-Leu or L-Glu and m approximately 3-6, i

Subject(s)
Glutamic Acid/chemistry , Leucine/chemistry , Oligopeptides/chemistry , Oligopeptides/chemical synthesis , Polylysine/chemistry , Serine/chemistry , Animals , Circular Dichroism , Mice , Oligopeptides/toxicity , Polylysine/analogs & derivatives , Protein Conformation , Protein Structure, Secondary , Solutions , Spleen/cytology , Spleen/drug effects
11.
Bioconjug Chem ; 10(5): 781-90, 1999.
Article in English | MEDLINE | ID: mdl-10502343

ABSTRACT

For the construction of macromolecule-drug conjugates, it is important to provide rational basis to the selection of proper carrier. With respect to the importance of the side-chain structure and charge of the branched polypeptides in biological properties, we have prepared a new class of branched polypeptides with single or multiple hydroxyl groups and studied their solution conformation, in vitro cytotoxicity, biodistribution, and immunoreactivity. For comparative studies, polypeptides were designed to contain serine at various positions of the side chains, varying also the number. Ser was attached to the end of oligo(DL-Ala) side chains grafted to polylysine resulting polypeptides with the general formula poly[Lys(Ser(i)-DL-Ala(m))], (SAK). Ser was also coupled directly to the polylysine backbone poly[Lys(Ser(i))] (S(i)K) and then elongated by polymerization of N-carboxy-DL-Ala anhydride resulting poly[Lys(DL-Ala(m)-Ser(i))] (ASK). An additional polymer was also prepared, but instead of the oligo(DL-Ala) branches, oligo(DL-Ser) side chains were introduced (poly[Lys(DL-Ser(m))], SK). The presence of hydroxyl groups resulted in compounds with improved of water solubility. CD spectra of polypeptides showed significant differences correlating with the position and numbers of Ser residues in the side chains. Under physiological conditions, polycationic polypeptides assumed ordered secondary structure (S(i)K and LSK) or partially unordered conformation (SK, SAK, and ASK). Data of selected polymers demonstrate that these polycationic compounds are essentially nontoxic in vitro on normal rat liver or mouse spleen cells and have no cytostatic effect on mouse colorectal carcinoma C26 cells. The blood clearance and biodistribution of these derivatives were greatly dependent on the position and number of Ser residues in the branches and possess a rather extended blood survival in mice. Polypeptides were taken up predominantly by the liver and kidney (S(i)K, LSK, and ASK) or kidney and lung (SK and SAK). The best survival in the blood was found with SAK, representing the first polycationic branched polypeptide, which show extended blood clearance. The relative position of Ser residue had also a marked influence on the immunogenicity of polypeptides. The characteristics of the antibody response to polypeptide containing Ser at the end of the branches (SAK) or adjacent to the polylysine backbone (ASK) was also dependent on the genetic background of the mouse strains. We also found that these compounds have no effect on to the SRBC-specific humoral immune response, indicating the lack of nonspecific immunostimulatory potential. In conclusion, these studies suggest that synthetic branched polypeptides with Ser can be considered as candidates for constructing suitable conjugates for drug/epitope delivery. It is not only due to the presence of hydroxyl group to be used for oxime chemistry but also to their beneficial biological features.


Subject(s)
Drug Carriers/chemical synthesis , Drug Carriers/toxicity , Peptides/chemical synthesis , Peptides/toxicity , Polyamines/chemical synthesis , Polyamines/toxicity , Adjuvants, Immunologic/pharmacology , Animals , Drug Carriers/metabolism , Female , Immunoglobulin G/biosynthesis , Immunoglobulin M/biosynthesis , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , Peptides/blood , Peptides/immunology , Polyamines/blood , Polyamines/immunology , Polyelectrolytes , Rats , Sheep , Solutions , Structure-Activity Relationship , Tissue Distribution , Tumor Cells, Cultured
12.
Proc Natl Acad Sci U S A ; 96(5): 2361-6, 1999 Mar 02.
Article in English | MEDLINE | ID: mdl-10051647

ABSTRACT

Conjugation of gonadotropin-releasing hormone (GnRH) analogues GnRH-III, MI-1544, and MI-1892 through lysyl side chains and a tetrapeptide spacer, Gly-Phe-Leu-Gly (X) to a copolymer, poly(N-vinylpyrrolidone-co-maleic acid) (P) caused increased antiproliferative activity toward MCF-7 and MDA-MB-231 breast, PC3 and LNCaP prostate, and Ishikawa endometrial cancer cell lines in culture and against tumor development by xenografts of the breast cancer cells in immunodeficient mice. MCF-7 cells treated with P-X-1544 and P-X-1892 displayed characteristic signs of apoptosis, including vacuoles in the cytoplasm, rounding up, apoptotic bodies, bleb formation, and DNA fragmentation. Conjugates, but not free peptides, inhibited cdc25 phosphatase and caused accumulation of Ishikawa and PC3 cells in the G2/M phase of the cell cycle after 24 h at lower doses and in the G1 and G2 phases after 48 h. Since P-X-peptides appear to be internalized, the increased cytotoxicity of the conjugates is attributed to protection of peptides from proteolysis, enhanced interaction of the peptides with the GnRH receptors, and/or internalization of P-X-peptide receptor complexes so that P can exert toxic effects inside, possibly by inhibiting enzymes involved in the cell cycle. The additional specificity of P-X-peptides compared with free peptides for direct antiproliferative effects on the cancer cells but not for interactions in the pituitary indicates the therapeutic potential of the conjugates.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Gonadotropin-Releasing Hormone/analogs & derivatives , Gonadotropin-Releasing Hormone/therapeutic use , Hormone Antagonists/therapeutic use , Animals , Apoptosis , Bone Marrow Transplantation/immunology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Cycle/drug effects , Cell Cycle Proteins/metabolism , Cell Division , DNA Fragmentation , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Female , Humans , Immunosuppression Therapy/methods , Maleates/therapeutic use , Mice , Mice, Inbred CBA , Phosphoprotein Phosphatases/metabolism , Polyvinyls/therapeutic use , Thymectomy , Transplantation, Heterologous , Transplantation, Isogeneic , Tumor Cells, Cultured , Whole-Body Irradiation , cdc25 Phosphatases
13.
Anticancer Res ; 19(4B): 3265-8, 1999.
Article in English | MEDLINE | ID: mdl-10652622

ABSTRACT

The somatostatin analog TT-232 containing a 5 residue ring, was previously shown to inhibit the proliferation of a large number of cancer cell lines in vitro and reduce the size of tumors in animal models in vivo. Its action is accompanied by inhibition of tyrosine kinases and is characterized by the induction of programmed cell death. On the other hand, it was proved to be free of the endocrine effects of the natural compound. The aim of this study was to find the optimal dose and administration route for in vivo tumor therapy in an animal model. We have investigated the dose--and administration route-dependent antitumor activity of TT-232 on S-180 sarcoma tumor transplanted to inbred BDF1 mice from SPF breeding. Long-term administration (i.p., s.c. and i.v. injections) was started either on the day subsequent to tumor transplantation or after the development of tumor. The antineoplastic potential of TT-232 was evaluated on the basis of survival and tumor growth inhibition. In long-term administration (injections twice a day for 2 weeks) a significant, but dose- and administration route-dependent therapeutic efficacy of TT-232 was observed. The optimum dose of TT-232 15 micrograms/kg which resulted in a 30-40% cure rate and 50-70% growth inhibition in S-180 sarcoma tumor. A moderate antitumor effect was achieved by TT-232 when it was administered after the evelopment of tumor. Our study suggests that TT-232 can be a promising antitumor agent.


Subject(s)
Antineoplastic Agents/therapeutic use , Peptides, Cyclic/therapeutic use , Sarcoma, Experimental/drug therapy , Animals , Antineoplastic Agents/administration & dosage , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Mice , Neoplasm Transplantation , Peptides, Cyclic/administration & dosage , Somatostatin/analogs & derivatives , Survival Analysis
14.
Eur J Cancer ; 34(1): 155-61, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9624251

ABSTRACT

The acid labile derivative of Daunomycin cis-aconityl Daunomycin (cAD), was coupled to an amphoteric polypeptide, poly[Lys-(Glui-DL-Alam)] (EAK), which was selected for conjugation on the basis of its pharmacological and immunological properties. The systemic toxicity of covalently attached Daunomycin was studied by monitoring body weight, life-span, bone marrow and haematological parameters of BDF1 mice. More than 3-fold the lethal dose of free Daunomycin could be applied without serious toxic effect when the drug was attached to EAK. The dose- and time-dependent modulatory effect of free drug and [cAD]-EAK conjugate on the humoral and cellular immune response to sheep red blood cell antigens in mice was studied. The conjugation of Daunomycin to EAK carrier polypeptide compensated for the immunosuppression induced by free Daunomycin. [cAD]-EAK conjugate at Daunomycin doses of 2-10 mg/kg was very effective against L1210 leukaemia producing 66-100% long-term survivors (> 60 days), while Daunomycin in itself increased the mean survival only by 52%, with no long-term survivors. The mixture of free Daunomycin and EAK polypeptide had similar toxicity and antitumour activity as free Daunomycin, indicating the important role of covalent attachment in increased therapeutic efficacy.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Daunorubicin/therapeutic use , Immunotoxins/therapeutic use , Peptides/metabolism , Animals , Antibiotics, Antineoplastic/adverse effects , Antibiotics, Antineoplastic/metabolism , Daunorubicin/adverse effects , Daunorubicin/metabolism , Female , Immunotoxins/adverse effects , Immunotoxins/metabolism , Intercellular Signaling Peptides and Proteins , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA
15.
Leukemia ; 11(10): 1769-74, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9324299

ABSTRACT

It has previously been reported that the use of mitobronitol (dibromomannitol, DBM) instead of busulphan (BU) for myelosuppression is associated with significantly decreased risk for several complications of allogeneic bone marrow transplantation in accelerated chronic granulocytic leukemia. In exploring the pharmacologic basis for this observation, we have compared the acute and subacute cytotoxicities of DBM and BU on the spleen and thymus of mice. While there was comparable early (day 3) weight loss in both organs following these treatments, splenic B cells exhibited significantly less damage, and thymic regeneration (over weeks) was significantly faster following DBM treatment than with BU. These observations raise the possibility that improved post-BMT immune recovery could contribute to the clinical benefits observed with DBM-preconditioning.


Subject(s)
Antineoplastic Agents, Alkylating/toxicity , B-Lymphocytes/drug effects , Bone Marrow Purging/adverse effects , Bone Marrow Transplantation/adverse effects , Busulfan/toxicity , Mitobronitol/toxicity , Thymus Gland/drug effects , Animals , Female , Mice , Mice, Inbred BALB C , Risk Factors , Spleen/cytology , Spleen/drug effects
16.
Proc Natl Acad Sci U S A ; 93(22): 12513-8, 1996 Oct 29.
Article in English | MEDLINE | ID: mdl-8901613

ABSTRACT

We report a series of new in vitro and in vivo data proving the selective antitumor activity of our somatostatin structural derivative, TT-232. In vitro, it inhibited the proliferation of 20 different human tumor cell lines in the range of 50-95% and induced a very strong apoptosis. In vivo TT-232 was effective on transplanted animal tumors (Colon 26, B16 melanoma, and S180 sarcoma) and on human tumor xenografts. Treatment of MDA-MB-231 human breast cancer xenografted in mice with low submaximal doses of TT-232 [0.25 and 0.5 mg/kg of body weight (b.w.)] caused an average 80% decrease in the tumor volume resulting in 30% tumor-free animals surviving for longer than 200 days. Treatment of prostate tumor (PC-3) xenografted animals with 20 mg/kg of b.w. of TT-232 for 3 weeks resulted in 60% decrease in tumor volume and 100% survival even after 60 days, while 80% of nontreated animals perished. We have demonstrated that TT-232 did not bind to the membrane preparation of rat pituitary and cortex and had no antisecretory activity. TT-232 was not toxic at a dose of 120 mg/kg of b.w. in mice. Long-term incubation (24 h) of tumor cells with TT-232 caused significant inhibition of tyrosine kinases in good correlation with the apoptosis-inducing effect. The level of p53 or KU86 did not change following TT-232 treatment, suggesting a p53-independent apoptotic effect. Preincubation of human breast cancer cells (MDA-MB-453) with TT-232 for 2 h decreased the growth factor receptor autophosphorylation. All of these data suggest that TT-232 is a promising and selective antitumor agent.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Peptides, Cyclic/pharmacology , Animals , Cell Division/drug effects , Dose-Response Relationship, Drug , Humans , Mice , Mice, Inbred BALB C , Protein-Tyrosine Kinases/metabolism , Rats , Somatostatin/analogs & derivatives , Tumor Cells, Cultured/drug effects
17.
Cancer Detect Prev ; 20(2): 153-9, 1996.
Article in English | MEDLINE | ID: mdl-8706041

ABSTRACT

The aim of the study was to test in vivo the gonadotropin-releasing hormone (GnRH) antagonists and their conjugates showing antitumor activities in vitro. The in vivo experiments with the human GnRH antagonist MI-1544 (Ac-D-Trp1,3,D-Cpa2,D-Lys6,D-Ala10)-GnRH, the chicken GnRH antagonist MI-1892 (Ac-D-Trp1,3,D-Cpa2,Lys5,/beta-Asp(DEA)/6,Gln8,D-Al a10)-GnRH, and their copolymer conjugates were carried out on MCF-7 and MDA-MB-231 human breast tumors xenografted in immunosuppressed CBA/Ca HRIJ-T6 female mice and on MXT mouse mammary tumors in BDF1 mice. The P-X-1544 and P-X-1892 conjugates were prepared by coupling the GnRH antagonists to macromolecule copolymer through biodegradable spacers. MI-1544 and its conjugate had strong, whereas MI-1892 and its conjugate had slight, castration effect in rats. All of them showed selective antitumor activity. The conjugates, given daily, inhibited both types of xenografts by 42 to 49%. Their activity was stronger in MXT mammary tumor (72 to 61%). The in vivo effect of GnRH antagonists was largely increased by coupling them to nonbiodegradable macromolecule carriers of polyanionic character. P-X-1544 and P-X-1892 GnRH antagonist-macromolecule conjugates might become important therapeutic agents for the treatment of breast cancer.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Gonadotropin-Releasing Hormone/analogs & derivatives , Animals , Disease Progression , Drug Carriers , Female , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Gonadotropin-Releasing Hormone/pharmacology , Gonadotropin-Releasing Hormone/therapeutic use , Hormone Antagonists/therapeutic use , Humans , Mammary Neoplasms, Experimental , Mice , Mice, Inbred CBA , Neoplasm Transplantation , Thymectomy , Transplantation, Heterologous
18.
Ann Nucl Med ; 9(4): 247-51, 1995 Nov.
Article in English | MEDLINE | ID: mdl-8770294

ABSTRACT

Radiolabelled synthetic branched chain polypeptides (BCP) represent a new and novel range of materials with potential as radiopharmaceuticals. Preliminary imaging studies have been undertaken with 111In-labelled BCP in mice with subcutaneously transplanted mammary carcinoma. Four polypeptides each with a poly(L-lysine) backbone and side chains of DL-alanine residues were studied. These were AK, which is polycationic, EAK which is amphoteric, having additional glutamic acid residues at the end of the side chains, and AcEAK (anionic) and SucEAK (highly polyanionic) where the terminal glutamic acid amino groups were acetylated or succinylated respectively. Radiolabelling was achieved by previous conjugation with DTPA. Serial images up to 48 hours showed marked retention of 111In-labelled polycationic AK and polyanionic SucEAK in the liver and spleen, with renal uptake also being visible in the case of AK. 111In-labelled EAK and AcEAK showed longer blood survival with some liver uptake, but tumour uptake was also visualized by 24 hours with both of these polypeptides. These studies demonstrate the feasibility of using 111In-labelled synthetic branched chain polypeptides as radiopharmaceuticals for gamma scintigraphy and the visualization of tumours by modification of the side chain structure. These materials warrant further study.


Subject(s)
Indium Radioisotopes , Mammary Neoplasms, Experimental/diagnostic imaging , Polylysine/analogs & derivatives , Animals , Electrochemistry , Female , Indium Radioisotopes/pharmacokinetics , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Inbred BALB C , Peptides/chemistry , Peptides/pharmacokinetics , Polylysine/chemistry , Polylysine/pharmacokinetics , Radionuclide Imaging , Tissue Distribution
19.
Anesth Analg ; 79(6): 1102-6, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7978432

ABSTRACT

Intravenous (IV) propofol was compared with IV thiopental/pentobarbital as a sedative for children undergoing magnetic resonance imaging (MRI) of the brain or spine. Fifty-eight outpatients (aged 11 mo to 6 1/2 yr, ASA grade I and II) were enrolled in the study and randomized to two groups. After IV cannulation, Group I received IV propofol (1-2 mg/kg), followed immediately by a propofol infusion (75-100 micrograms.kg-1.min-1). Group II received IV thiopental (1-3 mg/kg) followed by a pentobarbital bolus (2-3 mg/kg). Supplemental thiopental doses (1-2 mg/kg) were administrated to maintain adequate sedation. Discharge time and postanesthesia recovery scores were determined by an independent blinded observer. Time of recovery to full consciousness in Group I was significantly less than in Group II (19 +/- 7 min vs 35 +/- 20; P < 0.005). Time to discharge was also significantly less in Group I (24 +/- 6 min vs 40 +/- 11; P < 0.05). A preliminary cost analysis was applied to the clinical data obtained and to a theoretical model of a pediatric MRI center. Cost analysis of anesthesia services revealed added drug costs ($1600.76 per year for the propofol group) but significant savings of postanesthesia care unit (PACU) nursing time ($5086.67 per year). Outcomes such as patient morbidity and technical quality of the MRI scans did not differ significantly between the two groups. In conclusion, analysis of the clinical data suggests that propofol may be more suitable than barbiturates for children undergoing outpatient procedures despite its higher price.


Subject(s)
Anesthesiology/economics , Pentobarbital/therapeutic use , Propofol/therapeutic use , Thiopental/therapeutic use , Anesthesia Recovery Period , Child , Child, Preschool , Consciousness/drug effects , Drug Therapy/economics , Humans , Infant , Infusions, Intravenous , Magnetic Resonance Imaging/adverse effects , Magnetic Resonance Imaging/economics , Magnetic Resonance Imaging/methods , Pentobarbital/adverse effects , Propofol/adverse effects , Thiopental/adverse effects
20.
J Thorac Cardiovasc Surg ; 107(1): 280-8, 1994 Jan.
Article in English | MEDLINE | ID: mdl-8283897

ABSTRACT

Cardiopulmonary bypass has been shown in adults to activate platelets and leukocytes, lead to the formation of circulating platelet-leukocyte conjugates, and alter adhesive receptors on both cell types. Pediatric patients with congenital heart disease undergoing cardiopulmonary bypass, however, have not been extensively studied and may represent a group at particular clinical risk for bleeding and pulmonary dysfunction. We studied 13 patients with congenital heart disease undergoing operations necessitating bypass, 7 with cyanotic and 6 with noncyanotic congenital heart disease. We determined that (1) the surface density of platelet glycoprotein Ib was significantly lower at baseline and throughout bypass in patients with cyanotic heart disease than in noncyanotic patients; (2) platelet glycoprotein Ib in both cyanotic and noncyanotic congenital heart disease decreased significantly during bypass, with a nadir of 75% of baseline values; (3) platelets were activated to a high degree, comparable with that seen in adults; (4) mean circulating monocyte-platelet conjugates rose significantly during bypass, increasing from 36% to 66% by the end of bypass, whereas neutrophil-platelet conjugates and lymphocyte-platelet conjugates declined; and (5) both monocytes and neutrophils were activated by cardiopulmonary bypass, as assessed by increased surface expression of CD11b and, in the case of monocytes, CD11b expression continued to increase even after termination of bypass. Patients with cyanotic and noncyanotic heart disease did not differ with respect to platelet or leukocyte activation or the formation of platelet-leukocyte conjugates. We conclude that in children with congenital heart disease cardiopulmonary bypass causes loss of platelet adhesion receptors, activation of platelets, formation of platelet-leukocyte conjugates, and leukocyte activation. Cyanotic and noncyanotic patients are qualitatively similarly affected; however, cyanotic patients demonstrate a baseline deficit in the platelet adhesion receptor glycoprotein Ib. These cellular changes may contribute to both the hemostatic and inflammatory complications associated with cardiopulmonary bypass.


Subject(s)
Blood Platelets/physiology , Cardiopulmonary Bypass , Heart Defects, Congenital/surgery , Leukocytes/physiology , Cell Adhesion , Child, Preschool , Heart Defects, Congenital/blood , Humans , Infant , Infant, Newborn , Platelet Activation , Platelet Membrane Glycoproteins/metabolism , Receptors, Leukocyte-Adhesion/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...