Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Biomedicines ; 10(9)2022 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-36140319

RESUMO

High-grade serous ovarian carcinoma (HGSOC) is the most common type of epithelial ovarian cancer. The majority of cases are diagnosed at advanced stages, when intraperitoneal (IP) spread has already occurred. Despite significant surgical and chemotherapeutic advances in HGSOC treatment over the past decades, survival rates with HGSOC have only modestly improved. Chimeric antigen receptor (CAR)-T cells enable T cells to directly bind to tumor-associated antigens in a major histocompatibility complex-independent manner, thereby inducing tumor rejection. While CAR-T cell therapy shows great promise in hematological malignancies, its use in solid tumors is limited. Therefore, innovative approaches are needed to increase the specificity of CAR-modified T cells against solid tumors. The aim of this study was to assess the efficacy and safety of intraperitoneal (IP) versus intravenous (IV) CAR-T cell therapy in the treatment of HGSOC. We constructed a CAR that targets the ErbB2/HER2 protein (ErbB2CAR), which is overexpressed in HGSOC, and evaluated the functionality of ErbB2CAR on ovarian cancer cell lines (OVCAR8, SKOV3, and NAR). Our findings show that an IP injection of ErbB2CAR-T cells to tumor-bearing mice led to disease remission and increased survival compared to the IV route. Moreover, we found that IP-injected ErbB2CART cells circulate to a lesser extent, making them safer for non-tumor tissues than IV-injected cells. Further supporting our findings, we show that the effect of ErbB2CAR-T cells on primary HGSOC tumors is correlated with ErbB2 expression. Together, these data demonstrate the advantages of an IP administration of CAR-T cells over IV administration, offering not only a safer strategy but also the potential for counteracting the effect of ErbB2CAR in HGSOC. Significance: IP-injected ErbB2CAR-T cells led to disease remission and increased survival compared to the IV route. These findings demonstrate the advantages of IP administration, offering a safe treatment strategy with the potential for counteracting the effect of ErbB2CAR in HGSOC.

2.
Expert Rev Gastroenterol Hepatol ; 16(9): 851-861, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-36107723

RESUMO

INTRODUCTION: Peritoneal carcinomatosis (PC) is an advanced malignancy that is not sensitive to systemic conventional chemotherapy. Treatment options for PC are usually palliative rather than curative. Cytoreductive surgery and hyperthermic intraperitoneal (IP) chemotherapy are associated with limited efficacy in patients with PC. However, the peritoneum can produce effective immunity by inducing T-lymphocyte recruitment and proliferation, and the unique immune environment of the peritoneum provides the rationale for IP immunotherapy in PC. AREAS COVERED: The authors retrieved relevant documents of IP immunotherapy for PC from PubMed and Medline. This review elaborates on the knowledge of the peritoneal immune microenvironment and IP immunotherapy for PC covering immune stimulators, radioimmunotherapy, catumaxomab, cancer vaccines, chimeric antigen receptor (CAR)-T cells, and immune checkpoint inhibitors. EXPERT OPINION: The prognosis of PC is poor. However, the peritoneal cavity is a unique immune compartment with abundant immune cells which can produce effective immunity. IP immunotherapy may be a promising strategy in patients with PC.


Assuntos
Vacinas Anticâncer , Imunoterapia , Neoplasias Peritoneais , Receptores de Antígenos Quiméricos , Humanos , Vacinas Anticâncer/uso terapêutico , Inibidores de Checkpoint Imunológico/uso terapêutico , Imunoterapia/efeitos adversos , Neoplasias Peritoneais/terapia , Receptores de Antígenos Quiméricos/uso terapêutico , Microambiente Tumoral
3.
J Int Med Res ; 49(2): 300060520986664, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33616416

RESUMO

OBJECTIVE: This prospective, dose-escalation phase I study evaluated the safety and efficacy of intraperitoneal bevacizumab in managing refractory malignant ascites and explored the recommended dose of bevacizumab for further study. METHODS: Patients with refractory malignant ascites were enrolled. Bevacizumab was intraperitoneal administered weekly at an initial dose of 2.5 mg/kg, with dose escalation to 5 and 7.5 mg/kg performed following the standard "3 + 3" rule. The total duration of treatment was 2 or 3 weeks. RESULTS: Between December 2013 and September 2014, 13 patients (2.5 mg/kg, n = 4; 5 mg/kg, n = 3; 7.5 mg/kg, n = 6) with refractory malignant ascites were enrolled. Bevacizumab was well tolerated, and the most common treatment-related adverse events were abdominal pain (5/13), abdominal distension (2/13), and fatigue (2/13). The dose-limiting toxicity at 7.5 mg/kg was grade 3 bowel obstruction (1/13). The maximum tolerated dose (MTD) was not reached. The overall response and disease control rates were 7.7 and 61.5%, respectively. CONCLUSIONS: Intraperitoneal bevacizumab safe and well tolerated for treating malignant ascites, and the MTD was not reached at doses of 2.5 to 7.5 mg/kg. Intraperitoneal bevacizumab at 7.5 mg/kg weekly is recommended for further study to verify its anti-tumor activity.Trial registration: Clinical Trials NCT01852409.


Assuntos
Ascite , Neoplasias Peritoneais , Protocolos de Quimioterapia Combinada Antineoplásica , Ascite/tratamento farmacológico , Bevacizumab , Humanos , Dose Máxima Tolerável , Estudos Prospectivos
4.
Mol Ther Nucleic Acids ; 22: 791-802, 2020 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-33230476

RESUMO

Patients with peritoneal metastasis of gastric cancer have dismal prognosis, mainly because of inefficient systemic delivery of drugs to peritoneal tumors. We aimed to develop an intraperitoneal treatment strategy using amido-bridged nucleic acid (AmNA)-modified antisense oligonucleotides (ASOs) targeting synaptotagmin XIII (SYT13) and to identify the function of SYT13 in gastric cancer cells. We screened 71 candidate oligonucleotide sequences according to SYT13-knockdown efficacy, in vitro activity, and off-target effects. We evaluated the effects of SYT13 knockdown on cellular functions and signaling pathways, as well as the effects of intraperitoneal administration to mice of AmNA-modified anti-SYT13 ASOs. We selected the ASOs (designated hSYT13-4378 and hSYT13-4733) with the highest knockdown efficiencies and lowest off-target effects and determined their abilities to inhibit cellular functions associated with the metastatic potential of gastric cancer cells. We found that SYT13 interfered with focal adhesion kinase (FAK)-mediated intracellular signals. Intraperitoneal administration of hSYT13-4378 and hSYT13-4733 in a mouse xenograft model of metastasis inhibited the formation of peritoneal nodules and significantly increased survival. Reversible, dose- and sequence-dependent liver damage was induced by ASO treatment without causing abnormal morphological and histological changes in the brain. Intra-abdominal administration of AmNA-modified anti-SYT13 ASOs represents a promising strategy for treating peritoneal metastasis of gastric cancer.

5.
Int J Nanomedicine ; 13: 8063-8074, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30555227

RESUMO

BACKGROUND: PARP inhibitors, such as Olaparib, have advanced the treatment of ovarian cancer by providing patients with an effective and molecularly-targeted maintenance therapy. However, all orally-administered drugs, including Olaparib, must undergo first-pass metabolism. In contrast, a nanoparticle delivery system has the advantage of administering Olaparib directly into the peritoneal cavity for local treatment. Consequently, we sought to optimize the sustained-release formulation NanoOlaparib, previously deemed effective as an intravenous solid tumor treatment, for the local treatment of disseminated disease via intraperitoneal (i.p.) therapy. METHODS: The tumor cell line 404, which was derived from a Brca2 -/-, Tp53 -/-, Pten -/- genetically engineered mouse model, exhibited high sensitivity to Olaparib in vitro. It was chosen for use in developing an i.p. spread xenograft for testing nanotherapy efficacy in vivo. NanoOlaparib as a monotherapy or in combination with cisplatin was compared to oral Olaparib alone or in combination using two different dose schedules. A pilot biodistribution study was performed to determine drug accumulation in various organs following i.p. administration. RESULTS: Daily administration of NanoOlaparib reduced tumor growth and decreased the variability of the treatment response observed with daily oral Olaparib administration. However, systemic toxicity was observed in both the NanoOlaparib and vehicle (empty nanoparticle) treated groups. Scaling back the administration to twice weekly was well tolerated up to 100 mg/kg but reduced the effect on tumor growth. Biodistribution profiles indicated that NanoOlaparib began accumulating in tissues within an hour of administration and persisted for at least 72 hours after a single dose, exiting the peritoneal cavity faster than expected. CONCLUSION: NanoOlaparib must be modified for use against disseminated disease. Future avenues to develop NanoOlaparib as an i.p. therapy include a modified surface-coating to retain it in the peritoneal cavity and prevent entry into systemic circulation, in addition to targeting moieties for localization in tumor cells.


Assuntos
Antineoplásicos/administração & dosagem , Proliferação de Células/efeitos dos fármacos , Nanopartículas/administração & dosagem , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Peritoneais/tratamento farmacológico , Ftalazinas/administração & dosagem , Piperazinas/administração & dosagem , Animais , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Proteína BRCA2/fisiologia , Feminino , Humanos , Injeções Intraperitoneais , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Terapia de Alvo Molecular , Nanopartículas/química , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , PTEN Fosfo-Hidrolase/fisiologia , Neoplasias Peritoneais/metabolismo , Neoplasias Peritoneais/secundário , Ftalazinas/farmacocinética , Ftalazinas/farmacologia , Piperazinas/farmacocinética , Piperazinas/farmacologia , Distribuição Tecidual , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/fisiologia
6.
Anticancer Res ; 36(12): 6541-6546, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27919980

RESUMO

BACKGROUND: Our aim was to analyze the impact of intraperitoneal chemotherapy (IPC), administered with direct peritoneal puncture, on the survival of patients with pretreated ovarian cancer in a real-life setting. PATIENTS AND METHODS: This was a retrospective study comparing patients with advanced ovarian cancer treated with IPC (N=33) and patients treated with standard intravenous (i.v.) chemotherapy matching cases for known prognostic factors (age, platinum sensitivity, histological subgroup and grade). Data were then analyzed for survival with nested Cox multivariate regression. RESULTS: The case matching resulted in two homogeneous cohorts by age, platinum sensitivity, resistance to therapy and histology. When analyzed by hazard ratio (HR), the number of previous treatments and IPC vs. i.v. therapy were significant (HR=1.97 for i.v. and HR=1.90 for each incremental previous treatment line, multivariate p<0.001). When analyzing the patients with fewer than three previous treatment lines, IPC conferred a survival advantage of about 2.2 months (IPC=10.0 vs. i.v.=7.8 months, p=0.011). However, the survival advantage in heavily pre-treated patients (with three or more previous treatments) was not significant. One case, pre-treated with more lines of chemotherapy, with renal failure after intraperitoneal cisplatin was followed by death. None of the patients had bowel sub-occlusions and we recorded a lower incidence of local toxicity, such as cellulite, with IPC (two out of 33 cases). Two patients thereafter refused IPC due to abdominal pain. CONCLUSION: Our findings confirm that IPC is an effective approach compared to systemic chemotherapy for advanced ovarian cancer, even in pre-treated patients, including platinum-resistant cases. The survival benefit appears to be confined to non-heavily treated patients. Overall, direct intraperitoneal drug injection (without permanent devices) appears to be feasible, safe and possibly advantageous.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Neoplasias Ovarianas/tratamento farmacológico , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Cisplatino/administração & dosagem , Feminino , Humanos , Infusões Parenterais , Pessoa de Meia-Idade , Paclitaxel/administração & dosagem , Estudos Retrospectivos
7.
Oncotarget ; 7(37): 59322-59335, 2016 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-27528037

RESUMO

Ovarian cancer carries a significant mortality. Since symptoms tend to be minimal, the disease is often diagnosed when peritoneal metastases are already present. The standard of care in advanced ovarian cancer consists of platinum-based chemotherapy combined with cytoreductive surgery. Unfortunately, even after optimal cytoreduction and adjuvant chemotherapy, most patients with stage III disease will develop a recurrence. Intraperitoneal administration of chemotherapy is an alternative treatment for patients with localized disease. The pharmacological and physiochemical properties of melflufen, a peptidase potentiated alkylator, raised the hypothesis that this drug could be useful in ovarian cancer and particularily against peritoneal carcinomatosis. In this study the preclinical effects of melflufen were investigated in different ovarian cancer models. Melflufen was active against ovarian cancer cell lines, primary cultures of patient-derived ovarian cancer cells, and inhibited the growth of subcutaneous A2780 ovarian cancer xenografts alone and when combined with gemcitabine or liposomal doxorubicin when administered intravenously. In addition, an intra- and subperitoneal xenograft model showed activity of intraperitoneal administered melflufen for peritoneal carcinomatosis, with minimal side effects and modest systemic exposure. In conclusion, results from this study support further investigations of melflufen for the treatment of peritoneal carcinomatosis from ovarian cancer, both for intravenous and intraperitoneal administration.


Assuntos
Melfalan/análogos & derivados , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Peritoneais/tratamento farmacológico , Fenilalanina/análogos & derivados , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Linhagem Celular Tumoral , Procedimentos Cirúrgicos de Citorredução , Intervalo Livre de Doença , Avaliação Pré-Clínica de Medicamentos , Feminino , Humanos , Hipertermia Induzida , Injeções Intraperitoneais , Melfalan/uso terapêutico , Camundongos , Camundongos SCID , Recidiva Local de Neoplasia , Estadiamento de Neoplasias , Neoplasias Ovarianas/patologia , Neoplasias Peritoneais/patologia , Fenilalanina/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Pleura Peritoneum ; 1(4): 209-215, 2016 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-30911625

RESUMO

BACKGROUND: Peritoneal carcinomatosis in colorectal cancer is an advanced stage of the disease where improved survival can be attained whenever the resection associated with hyperthermic intreperitoneal chemotherapy is possible. In unresectable cases, systemic chemotherapy is administered to obtain conversion to resectability but results have not yet been clearly evaluated. Local chemotherapy in this setting has been proven useful in several similar situations. The aim of the present pilot study was to evaluate the feasibility of pre-operative intraperitoneal chemotherapy with oxaliplatin in these patients. METHODS: Six patients with unresectable peritoneal disease of colorectal origin were included in the study. An intraperitoneal implantable chamber catheter was inserted during the laparotomy that evaluated the extent of the peritoneal disease (peritoneal carcinomatosis index 25 to 39). Patients then underwent intraperitoneal chemotherapy with oxaliplatin 85 mg/m2 in combination with systemic chemotherapy (FOLFIRI or simplified LV5FU) and a targeted therapy every 2 weeks. RESULTS: Two catheter perfusion incidents were reported due to the abdominal wall thickness. Two patients completed the four intraperitoneal (IP) chemotherapy cycles without major toxicity. One patient developed grade 3 or 4 diarrhea requiring a short intensive care unit (ICU) stay, though it is not clear whether the event was induced by intravenous irinotecan, IP oxaliplatin or the combination of both. Grade 3 fatigue and abdominal pain were also recorded. For one patient with aggressive disease, best supportive care was initiated after the first course of chemotherapy. CONCLUSIONS: Our study is the first to assess intraperitoneal oxaliplatin-based chemotherapy in the preoperative setting for patients with unresectable peritoneal metastases. The tolerance was acceptable for 85 mg/m2 IP oxaliplatin combined with systemic therapy in these patients. Our results justify carrying on with a phase I/II trial to determine the recommended dose of oxaliplatin in this clinical context and its efficacy.

9.
Braz. arch. biol. technol ; 53(3): 609-613, May-June 2010. graf, tab
Artigo em Inglês | LILACS | ID: lil-548581

RESUMO

This study aimed to investigate the effects of the administration of butanolic residue (BR) of Pfaffia paniculata by intraperitoneal route to Ehrlich ascitis tumor bearing mice. Initially, a toxicity study of P. paniculata BR was performed in which doses of 12.5; 25 and 50mg/Kg were administered by intraperitoneal injection for seven days to Swiss mice. The treatment did not show toxicity. Then, Swiss male mice received, by intraperitoneal injection, once a day, 12.5; 25 or 50mg/Kg of P. paniculata BR for seven days. This protocol started in the same day of tumor inoculation with 5X10(6) cells i.p. The treatment with butanolic residue of P.paniculata i.p caused a significant increase in the ascitic volume; however, a significant decrease in tumor cells number per ml (p<0.05) was observed in P. paniculata treated mice that was followed by a numerical (although non-significant) decrease in the total numbers of tumor cells in the collected ascitic fluid. These results indicated a tumor cell inhibitory effect by P. paniculata butanolic residue in this experimental system, and indicate that topical application of this residue can be useful to control the cancer growth.


Neste estudo, foi avaliado o efeito do tratamento intraperitoneal com Resíduo Butanólico de Pfaffia paniculata, sobre o crescimento do Tumor de Ehrlich, forma ascítica. Foram utilizados dois grupos de 15 camundongos cada, sendo um grupo controle e o outro grupo tratado com RB 50mg/Kg. Todos os animais foram inoculados intraperitonealmente, com 5X10(6) células tumorais O tratamento iniciou-se no mesmo dia da inoculação do tumor. Assim, os animais receberam diariamente, por via intraperitoneal, 0,1 ml de RB na concentrações 50 mg/Kg, ou PBS como controle. Após 7 dias da inoculação do tumor, os animais foram eutanasiados e foi colhido o fluído ascítico total, para a contagem do número de células tumorais presentes neste fluído e estudo da morfologia destas células . Neste experimento observou-se aumento significante da quantidade de fluido ascítico nos animais tratados com RB, e diminuição significativa em relação ao número de células tumorais/ml e células tumorais totais, presentes no fluído ascítico, comparativamente com os animais controle. Estes resultados sugerem efeito inibitório tópico do RB levando à morte as células neoplásicas.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...