Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Radiat Oncol J ; 41(1): 23-31, 2023 Mar.
Article in English | MEDLINE | ID: mdl-37013415

ABSTRACT

PURPOSE: To perform the analysis of the peripheral blood lymphocyte changes after stereotactic ablative radiotherapy (SABR) in patients with oligometastatic cancers. MATERIALS AND METHODS: The dynamics of the immune status in peripheral blood was prospectively evaluated in 46 patients with lung (17 cases) or liver (29 cases) metastases treated by SABR. Flow cytometry of peripheral blood lymphocyte subpopulations was performed before SABR, 3-4 weeks and 6-8 weeks after the end of SABR: 3 fractions of 15-20 Gy or 4 fractions of 13.5 Gy. The number of treated lesions varied from 1 (32 patients) to 2-3 (14 patients). RESULTS: SABR induced a significant increase of T-lymphocytes (CD3+CD19-) (p = 0.001), T-helper (CD3+CD4+) (p = 0.004), activated cytotoxic T-lymphocytes (CD3+CD8+HLA-DR+) (p = 0.001), activated T-helpers (CD3+CD4+HLA-DR+) (p < 0.001). A significant decrease of T-regulated immune suppressive lymphocytes (CD4+CD25brightCD127low) (p = 0.002) and NKT-cells (CD3+CD16+CD56+) (p = 0.007) was recorded after the SABR. The comparative analysis demonstrated that lower doses of SABR (EQD2Gy(α/ß=10) = 93.7-105.7 Gy) induced significant increase of T-lymphocytes, activated cytotoxic T-lymphocytes, and activated CD4+CD25+ T-helpers, while SABR with higher doses (EQD2Gy(α/ß=10) = 150 Gy) was not associated with these effects. A more efficient activations of T-lymphocytes (p = 0.010), activated T-helpers (p < 0.001), and cytotoxic T-lymphocytes (p = 0.003) were associated with SABR to a single lesion. A significant increase of T-lymphocytes (p = 0.002), T-helpers (p = 0.003), and activated cytotoxic T-lymphocytes (p = 0.001) was observed after SABR for hepatic metastases in contrast to SABR for lung lesions. CONCLUSION: Changes in peripheral blood lymphocytes after SABR could be influenced by the location or the number of irradiated metastasis, and the dose of SABR.

2.
Oncologist ; 26(5): 364-e734, 2021 05.
Article in English | MEDLINE | ID: mdl-33749049

ABSTRACT

LESSONS LEARNED: Melatonin did not increase the efficacy of systemic chemotherapy in melanoma. Metformin did not increase the efficacy of systemic chemotherapy in melanoma. BACKGROUND: Current data support the possibility of antitumor activity of melatonin and metformin. METHODS: From March 2014 to December 2016, 57 patients with disseminated melanoma received dacarbazine (DTIC) 1,000 mg/m2 on day 1 of a 28-day cycle, either as monotherapy (first group) or in combination with melatonin 3 mg p.o. daily (second group) or metformin 850 mg two times a day p.o. daily (third group) as the first-line of chemotherapy. The primary endpoint was objective response rate (ORR). Secondary endpoints were time to progression (TTP), overall survival (OS), immunologic biomarkers, and quality of life. RESULTS: ORR was 7% and did not differ among the treatment groups. Median TTP was 57, 57, and 47 days, respectively, in the first, second, and third groups (р = .362). Median OS was 236, 422, and 419 days, respectively (p = .712). Two patients from the combinations groups showed delayed response to therapy. The increase of CD3+ CD4+ HLA-DR+ lymphocytes (p = .003), CD3+ CD8+ HLA-DR+ (p = .045), CD3+ CD8+ lymphocytes (p = .012), CD4+ CD25high CD127low lymphocytes (p = .029), and overall quantity of lymphocytes (p = .021) was observed in patients with clinical benefit. CONCLUSION: No benefit was found in either combination over DTIC monotherapy. Delayed responses in melatonin and metformin combination groups were registered. The increase of lymphocyte subpopulations responsible for antitumor immune response demonstrates the immune system's potential involvement in clinical activity.


Subject(s)
Melanoma , Melatonin , Metformin , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Dacarbazine/therapeutic use , Humans , Melanoma/drug therapy , Melatonin/pharmacology , Melatonin/therapeutic use , Metformin/pharmacology , Metformin/therapeutic use , Quality of Life
3.
Chemotherapy ; 65(1-2): 42-50, 2020.
Article in English | MEDLINE | ID: mdl-32772021

ABSTRACT

INTRODUCTION: The effects of chemotherapy are known to depend on the time of administration. Circadian rhythms are disturbed in tumors and in tumor bearers. Agents involved in controlling the circadian rhythms (chronobiotics) potentially can modify the outcomes of chemotherapeutics administered at different times of the day. Pineal hormone melatonin (MT) is a prototypic chronobiotic. OBJECTIVE: The aim of the study was to investigate if MT can affect efficacy or toxicity of chemotherapy drugs administered at the extreme time points of the working day of hospital personnel. METHODS: Cyclophosphamide, adriamycin, and 5-fluorouracil (CAF) and adriamycin and docetaxel (AT) cytotoxic drug combinations were administered on day 0 at 11:00 a.m. or at 5:00 p.m. (UTC+03:00) to 6-month-old female HER2/neu transgenic FVB/N mice bearing mammary adenocarcinomas. Some mice were additionally provided with MT in drinking water (20 mg/L) at night 1 week before or 3 weeks after treatment or during both periods. Tumor node sizes, body weight, and blood cell counts were determined right before treatment and on days 2, 7, 14, and 21. RESULTS: Significant decrease in the mean tumor node volume was found by days 14 and 21 upon all CAF and AT treatment schedules, except in animals treated with AT at 5:00 p.m. without supplementation with MT. In the latter case, mean tumor node volume on day 21 was the same as in the control. Supplementation of AT administered at 5:00 p.m. with MT improved the tumor response. CAF and AT regimens supplemented with MT also augmented the number of tumor nodes that did not increase by more than 20% by day 21 as compared to CAF or AT alone, respectively. This effect was significant in groups treated with AT at 5:00 p.m. and consistent upon other schedules. On day 7, leukopenia and anemia were registered in groups treated with CAF regimen; however, blood cell counts normalized by day 14. Both CAF and AT were associated with drop in the body weight registered on day 7. Supplementation with MT did not affect changes of the body weight and blood counts. CONCLUSIONS: MT supplementation to cytotoxic drugs can improve antitumor response, especially if it is blunted because of an inappropriate time of administration.


Subject(s)
Antineoplastic Agents/therapeutic use , Leukopenia/etiology , Melatonin/administration & dosage , Receptor, ErbB-2/metabolism , Anemia/etiology , Animals , Antineoplastic Agents/adverse effects , Blood Cell Count , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cyclophosphamide/adverse effects , Cyclophosphamide/therapeutic use , Disease Models, Animal , Docetaxel/adverse effects , Docetaxel/therapeutic use , Doxorubicin/adverse effects , Doxorubicin/therapeutic use , Drug Therapy, Combination , Female , Fluorouracil/adverse effects , Fluorouracil/therapeutic use , Mice , Mice, Transgenic
4.
Oncologist ; 25(9): e1303-e1317, 2020 09.
Article in English | MEDLINE | ID: mdl-32240562

ABSTRACT

LESSONS LEARNED: This study showed that carefully selected patients with locally advanced and metastatic forms of malignant melanoma and renal cell carcinoma could potentially have long-term disease control with a tag-7 gene-modified tumor cells-based vaccine. Randomized clinical trials in patients whose tumors produce low amounts of immunosuppressive factors are needed to confirm this hypothesis in both the adjuvant and metastatic settings. BACKGROUND: Immunotherapy may produce long-lasting effects on survival and toxicity. The magnitude of efficacy may be dependent on immune factors. We analyzed the results of a phase I/II study of a tag-7 gene-modified tumor cells-based vaccine (GMV) in patients with malignant melanoma (MM) or renal cell carcinoma (RCC) with biomarker analysis of immunosuppressive factors (ISFs) production by their tumor cells. METHODS: From 2001 to 2014, 80 patients received GMV: 68 with MM and 12 with RCC. Treatment in the metastatic setting included 61 patients (MM, 51; RCC, 10), and treatment in the adjuvant setting (after complete cytoreduction) included 19 patients (MM, 17; RCC, 2). Twenty-six patients were stage III (33%), and 54 (67%) were stage IV. The patients' tumor samples were transferred to culture, transfected with tag-7 gene, and inactivated by radiation. The produced product was injected subcutaneously every 3 weeks until progression or 2 years of therapy. ISFs were measured in the supernatants of the tumor cell cultures and used as predictive factors. RESULTS: No major safety issues or grade 5 adverse events (AEs) were seen. One grade 4 and two grade 3 AEs were registered. No AEs were registered in 89.4% of treatment cycles. No delayed AE was found. The 5-year overall survival (OS) in the intention-to-treat population was 25.1%. There were no differences between MM OS and RCC OS (log rank, p = .44). Median OS in the metastatic setting was 0.7 years and in the adjuvant setting was 3.1 years. Classification trees were built on the basis of ISF production (Fig. 1). The median OS was 6.6 years in the favorable prognosis (FP) group (major histocompatibility complex class I polypeptide-related sequence A [MICA] level ≤582 pg/mL, n = 15) and 4.6 months in the unfavorable (UF) group (MICA level >582 pg/mL, n = 12; p < .0001). No significant differences were found between classification trees based on ISFs (transforming growth factor ß1 [TGF-ß1], interleukin-10 [IL-10], and vascular endothelial growth factor [VEGF]). In patients with stage III-IV MM with FP, median OS was 2.3 years, with 31% patients alive at 10 years (Fig. 2) in the UF group (0.4 years; log rank, p = 1.94E-5). No FP patients received modern immunotherapy. CONCLUSION: GMV showed high results in carefully selected patients with low ISF (TGF-ß1, IL-10, and VEGF) production. The method should be further investigated in patients with FP.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Melanoma , Vaccines , Carcinoma, Renal Cell/therapy , Humans , Kidney Neoplasms/therapy , Melanoma/therapy , Vascular Endothelial Growth Factor A
5.
Clin Sarcoma Res ; 10: 3, 2020.
Article in English | MEDLINE | ID: mdl-32042403

ABSTRACT

BACKGROUND: Autologous dendritic cells (DC) loaded with tumor-associated antigens (TAAs) are a promising approach for anticancer immunotherapy. Polyantigen lysates appear to be an excellent source of TAAs for loading onto the patient's dendritic cells. Cancer/testis antigens (CTA) are expressed by a wide range of tumors, but are minimally expressed on normal tissues, and could serve as a universal target for immunotherapy. However, CTA expression levels can vary significantly in patients with the same tumor type. We proposed that patients who do not respond to DC-based therapy may have distinct features of the CTA expression profile on tumor cells. PATIENTS AND METHODS: We compared the gene expression of the principal families CTA in 22 melanoma and 27 soft tissue and bone sarcomas cell lines (STBS), received from patients and used for DC vaccine preparation. RESULTS: The majority (47 of 49, 95.9%) cell lines showed CTA gene activity. The incidence of gene expression of GAGE, NYESO1, MAGEA1, PRAME's was significantly different (adj. p < 0.05) between melanoma and sarcoma cell lines. The expression of the SCP1 gene was detected neither in melanoma cells nor in the STBS cells. Clustering by the gene expression profile revealed four different expression patterns. We found three main patterns types: hyperexpression of multiple CTA, hyperexpression of one CTA with almost no expression of others, and no expression of CTA. All clusters types exist in melanoma and sarcoma cell lines. We observed dependence of killing efficacy from the PRAME (rho = 0.940, adj. p < 0.01) expression during real-time monitoring with the xCELLigence system of the interaction between melanoma or sarcoma cells with the T-lymphocytes activated by the lysate of selected allogenous melanoma cell lines with high expression of CTA. CONCLUSION: Our results demonstrate that one can use lysates from allogeneic melanoma cell lines as a source of CTA for DC load during the production of anticancer vaccines for the STBS treatment. Patterns of CTA expression should be evaluated as biomarkers of response in prospective clinical trials.

6.
Integr Cancer Ther ; 18: 1534735419833778, 2019.
Article in English | MEDLINE | ID: mdl-30841763

ABSTRACT

This study aimed to evaluate the effect of lignin-derived polyphenolic composition BP-C3 on the efficacy and hematological toxicity of cyclophosphamide (CPA). Male and female Swiss-H derived mice bearing benzo[a]pyrene-induced soft tissue sarcomas were treated with CPA 300 mg/kg, BP-C3 75 mg/kg, or a combination. Tumor growth inhibition in male mice treated with CPA, BP-C3, or a combination of CPA and BP-C3 was significant and corresponded to 78%, 45%, and 82%, respectively, on day 21 after CPA administration on day 0. In female mice, tumor growth inhibition was 58%, -11%, and 35% when treated with CPA, BP-C3, or a combination of CPA and BP-C3, respectively. CPA administration resulted in significant hematological toxicity evidenced by a decreased white blood cell count on day 4 (2.43 ± 1.77 × 109/L in male mice and 1.19 ± 0.71 × 109/L in female mice) and anemia development on day 7 (6.55 ± 1.74 × 1012/L in male mice and 5.89 ± 2.24 × 1012/L in female mice). The red blood cell count measured on day 7 in animals treated with the combination of BP-C3 and CPA constituted 7.12 ± 1.17 × 1012/L and 7.36 ± 2.07 × 1012/L for male and female mice, respectively. The results of our study demonstrate the antitumor activity of BP-C3 in male mice bearing soft tissue sarcomas. Neither the antitumor activity nor the hematological toxicity of CPA were significantly influenced by BP-C3. A less pronounced effect of CPA on RBC count is demonstrated when this agent is given jointly with BP-C3.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzo(a)pyrene/pharmacology , Cyclophosphamide/adverse effects , Cyclophosphamide/pharmacology , Hematologic Diseases/chemically induced , Polyphenols/pharmacology , Sarcoma/drug therapy , Animals , Female , Male , Mice
7.
Int J Radiat Biol ; 94(2): 114-123, 2018 02.
Article in English | MEDLINE | ID: mdl-29251085

ABSTRACT

PURPOSE: There remains an unmet medical need for radioprotective and mitigative agents. BP-C2 is a novel lignin-derived polyphenolic composition with ammonium molybdate, developed as radioprotector/radiomitigator. OBJECTIVES: The present study evaluated BP-C2 for the mitigation of acute radiation syndrome (ARS). METHODS: A total-body irradiation mouse model (TBI, 4.0-8.0 Gy) was used in the study. RESULTS: In a 30-day survival study, performed in CBA mice, BP-C2, at a dosage of 81.0 mg/kg, improved survival (dose reduction factor (DRF) = 1.1) and increased the formation of endogenous spleen colony-forming units (CFU). In C57BL/6 mice, BP-C2, when administered daily for 7 days, starting 24 hours after TBI, also improved survival. In animals irradiated with 5.0 Gy, BP-C2 increased the number of CFUs (6.7 ± 5.1) compared to the 5.0 Gy placebo group (2.3 ± 2.3, p = .0245). The number of surviving intestinal crypts was maintained in the 5.0 Gy BP-C2 group (133.7 ± 13.9), in contrast to the 5.0 Gy placebo group (124.2 ± 10.5, p < .0023). BP-C2 also increased the number of LGR5 + positive cells in intestinal crypts. CONCLUSION: BP-C2 mitigates radiation-induced damage in mid-lethal range of radiation doses. Effects are mediated by enhancement of extramedullar hematopoiesis in the spleen and a protective effect on the intestinal epithelium.


Subject(s)
Lignin/chemistry , Molybdenum/pharmacology , Polyphenols/chemistry , Radiation-Protective Agents/pharmacology , Whole-Body Irradiation/mortality , Acute Radiation Syndrome , Animals , Cell Proliferation , Cell Survival , Disease Models, Animal , Leukocytes, Mononuclear/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Stem Cells
8.
Oncotarget ; 8(59): 100951-100956, 2017 Nov 21.
Article in English | MEDLINE | ID: mdl-33400728

ABSTRACT

The conference "Results and prospects of development of new polyphenolic drugs for cancer patients" took place at the N.N. Petrov National Medical Research Center of Oncology (PNMRCO) on May 31, 2017, and gathered researchers involved in development and evaluation of medicinal products based on the novel lignin-derived soluble polyphenolic polymer BP-Cx-1. BP-Cx-1 is the platform for a portfolio of innovative pharmacological products such as BP-C1, BP-C2 and BP-C3.

9.
Aging (Albany NY) ; 8(9): 1866-1875, 2016 08 28.
Article in English | MEDLINE | ID: mdl-27574962

ABSTRACT

Effects of long-term application of novel polyphenolic composition BP-C3, containing polyphenolic benzenepolycarboxylic acids, vitamins and minerals on some biomarkers of aging, life span and spontaneous tumorigenesis has been studied in female SHR mice. Administration of BP-C3 with drinking water (0.005%) did not exert any toxic effect (did not have effect on general condition of animals, weight dynamics and consumption of food), postponed age-related switch-off of estrous function, caused slight reduction of body temperature. An increased survival was observed in mice treated with BP-C3 (p=0.00164, log rank test). BP-C3 increased mean lifespan - by 8.4%, lifespan of the last 10% of animals - by 12.4%, and life span of tumor-free mice - by 11.6%. A tendency in ability of BP-C3 to inhibit development of spontaneous tumors in mice was detected, though it did not reach the level of statistical significance (p=0.166, log rank test). The number of malignant mammary tumors was 1.5 times less and total number of tumors of various localizations was 1.6 times less in BP-C3 treated animals. Multiple tumors were registered in 8% of mice in the сontrol group and no cases - in BP-C3 treated group. Thus, BP-C3 demonstrated some anti-carcinogenic and a pronounced geroprotective activity.


Subject(s)
Aging/drug effects , Carcinogenesis/drug effects , Longevity/drug effects , Polyphenols/administration & dosage , Animals , Body Temperature/drug effects , Body Weight/drug effects , Cell Transformation, Neoplastic/drug effects , Estrous Cycle/drug effects , Female , Mice
10.
Target Oncol ; 11(2): 235-8, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26264150

ABSTRACT

Vemurafenib, a specific inhibitor of mutated BRAF kinase, may activate wild-type BRAF and therefore induce squamous cell skin carcinomas in patients treated for melanoma. All vemurafenib clinical trials excluded patients with multiple primary malignant tumors; therefore, the action of this drug on concurrent BRAF wild-type malignancies remains insufficiently studied. We observed a patient, who was administered vemurafenib for BRAF mutation-containing melanoma, but experienced immediate relapse of previously controlled breast cancer disease. Interestingly, breast cancer lesions underwent regression soon after vemurafenib discontinuation. Therefore, caution must be taken while considering vemurafenib treatment for patients with multiple tumors.


Subject(s)
Antineoplastic Agents/adverse effects , Breast Neoplasms/pathology , Carcinoma, Squamous Cell/chemically induced , Indoles/adverse effects , Neoplasm Recurrence, Local/chemically induced , Sulfonamides/adverse effects , Breast Neoplasms/drug therapy , Disease Progression , Female , Humans , Indoles/therapeutic use , Melanoma/drug therapy , Melanoma/pathology , Middle Aged , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/metabolism , Sulfonamides/therapeutic use , Vemurafenib
11.
Adv Exp Med Biol ; 601: 387-93, 2007.
Article in English | MEDLINE | ID: mdl-17713028

ABSTRACT

Tumor growth is accompanied by active immune reactions even on the early stages. Vaccine therapy implies the use of single antigen or combination of antigens, either with or without adjuvants, for the modulation of immune response. N.N. Petrov Institute of Oncology joined the field of antitumor vaccine therapy and related cellular technologies in 1998. The following activities are held: (1) Optimization of the preparation of autologous and allogeneic antitumor vaccines and development of tumor cell culture bank for the experiments on allogeneic vaccination. (2) Clinical evaluation of autologous vaccine therapy by (a) bone marrow precursors of dendritic cells (DCs), which are loaded with tumor lysates; (b) genetically modified tumor cells; (c) intact tumor cells used in combination with various adjuvants (BCG, IL-1beta, and IL-1beta combined with low doses of cyclophosphamide) in patients with disseminated melanoma, metastatic kidney cancer, and colorectal cancer. Total 117 patients have received non-modified vaccine (48 patients: 2-6 intracutaneous BCG injections; 54 patients: 4-6 intracutaneous IL-1beta injections; 15 patients: up to 6 injections of IL-1beta in combination with low doses of cyclophosphamide). Clinical trial of genetically modified vaccine included 59 patients (clinical results: I PR (partial response) / 8 SD (disease stabilization)--melanoma, 2 PR/ 2 MR (minimal response) / 3 SD--renal cancer). Vaccine prepared from tumor cell-activated DC bone marrow precursors was administered to 18 patients (clinical results: 2 MR and 6 SD).


Subject(s)
Cancer Vaccines/chemistry , Immunotherapy/methods , Neoplasms/immunology , Neoplasms/therapy , Adult , Aged , Antigens, Neoplasm , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Dendritic Cells/immunology , Female , Humans , Immune System , Immunotherapy, Active , Male , Middle Aged , Models, Biological
SELECTION OF CITATIONS
SEARCH DETAIL
...