Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 132
Filtrar
2.
Appl Immunohistochem Mol Morphol ; 30(4): 264-272, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35384876

RESUMO

Secretory carcinoma (SC) is a low-grade salivary gland carcinoma characterized by recurrent ETV6 rearrangements. Most cases have ETV6-NTRK3 fusions, while the minority of cases have non-NTRK3 fusions, including ETV6-RET and ETV6-MET. Detection of the fusion partner has become important, as there are TRK or RET inhibitors that may benefit patients with advanced SC. Currently, there are different methods to detect gene rearrangement in SCs, such as next-generation sequencing, reverse transcription-polymerase chain reaction, or fluorescence in situ hybridization. Immunohistochemistry (IHC) has greater accessibility, quick turnaround time, and can serve as a screening tool for confirmatory molecular tests. Pan-TRK and RET antibodies have been used to detect gene fusions in different tumors. Here, pan-TRK and RET IHC assays were performed on 28 salivary gland SCs, including 27 cases with ETV6-NTRK3 and one with ETV6-RET fusion confirmed by fluorescence in situ hybridization. Pan-TRK staining was positive in 26/27 (96.3%) of NTRK3 fusion-positive SCs with a nuclear staining pattern in more than 50% of tumor cells, and negative in the RET-rearranged case. RET IHC showed positive staining in most cases (26/28), but only three cases (including the RET-rearranged case) had diffuse and strong staining. RET IHC can be considered an effective screening test when diffuse/strong reactivity is present in pan-TRK IHC-negative cases. This study showed that pan-TRK staining has high sensitivity and specificity for SC with NTRK3 fusion. Whereas pan-TRK IHC is a useful screening method, further studies are needed to assess the value of RET IHC as a second sequential step.


Assuntos
Adenocarcinoma , Carcinoma , Neoplasias das Glândulas Salivares , Biomarcadores Tumorais/genética , Carcinoma/diagnóstico , Fusão Gênica , Humanos , Imuno-Histoquímica/métodos , Hibridização in Situ Fluorescente , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/imunologia , Proteínas Proto-Oncogênicas c-ret/genética , Proteínas Proto-Oncogênicas c-ret/imunologia , Receptor trkA/genética , Receptor trkA/imunologia , Neoplasias das Glândulas Salivares/diagnóstico , Neoplasias das Glândulas Salivares/genética , Neoplasias das Glândulas Salivares/patologia , Glândulas Salivares/patologia
3.
Histopathology ; 80(4): 686-697, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34821406

RESUMO

AIMS: To evaluate the diagnostic accuracy of SSX and SSX::SS18 antibodies in decalcified surgical specimens and outcome of synovial sarcomas (SS) of bone. METHODS AND RESULTS: Twenty-five cases were classified as bone SS (prevalence 0.32% among malignant primary bone sarcoma). Median age was 34 years (range = 9-79). Twenty-four of 25 patients presented with non-metastatic tumours, one with lung metastases. The majority of tumours involved the long bones of extremities with metaphyseal origin. Mean size of the tumour was 7.1 cm. Twenty cases (80%) were monophasic and five (20%) were biphasic. SS18::SSX fusion-specific antibody had 92% sensitivity and 99% specificity for primary bone SS, whereas SSX C-terminus antibody had 100% sensitivity and 94% specificity. Fluorescence in-situ hybridisation (FISH) analysis was feasible in nine (36%) cases and detected SS18 rearrangement in all nine cases. All patients underwent surgical removal of their primary tumour, with adequate margins in 18 (72%) patients. Chemotherapy with metothrexate, cisplatin, doxorubicin and ifosfamide was used in the seven patients. Two patients with inadequate surgical margins received radiotherapy. With a median follow-up of 80 months (range = 6-428), 5- and 10-year overall survival (OS) was 66.6% and 47.9%, respectively, and 5 and 10 years' disease-free survival (DFS) was 36.8% [95% confidence interval (CI) = 18.0-55.7%] and 32.2% (95% CI = 14.6-51.2%), respectively. A significant improvement in 10 years' DFS in patients undergoing chemotherapy compared with patients who did not was observed (P = 0.039). CONCLUSIONS: Our series highlights the utility of SS18::SSX fusion-specific and SSX C-terminus antibodies to support the diagnosis of SS. Adjustment chemotherapy was associated with improved prognosis in this series.


Assuntos
Anticorpos/análise , Neoplasias Ósseas/diagnóstico , Proteínas de Fusão Oncogênica/imunologia , Sarcoma Sinovial/diagnóstico , Adolescente , Adulto , Idoso , Neoplasias Ósseas/patologia , Criança , Feminino , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Sarcoma Sinovial/patologia , Adulto Jovem
4.
Mod Pathol ; 34(4): 748-757, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33299109

RESUMO

Alveolar Rhabdomyosarcoma (ARMS) is an aggressive pediatric cancer with about 80% of cases characterized by either a t(1;13)(p36;q14) or t(2;13)(q35;q14), which results in the formation of the fusion oncogenes PAX7-FOXO1 and PAX3-FOXO1, respectively. Since patients with fusion-positive ARMS (FP-RMS) have a poor prognosis and are treated with an aggressive therapeutic regimen, correct classification is of clinical importance. Detection of the translocation by different molecular methods is used for diagnostics, including fluorescence in situ hybridization and RT-PCR or NGS based approaches. Since these methods are complex and time consuming, we developed specific monoclonal antibodies (mAbs) directed to the junction region on the PAX3-FOXO1 fusion protein. Two mAbs, PFM.1 and PFM.2, were developed and able to immunoprecipitate in vitro-translated PAX3-FOXO1 and cellular PAX3-FOXO1 from FP-RMS cells. Furthermore, the mAbs recognized a 105 kDa band in PAX3-FOXO1-transfected cells and in FP-RMS cell lines. The mAbs did not recognize proteins in fusion-negative embryonal rhabdomyosarcoma cell lines, nor did they recognize PAX3 or FOXO1 alone when compared to anti-PAX3 and anti-FOXO1 antibodies. We next evaluated the ability of mAb PFM.2 to detect the fusion protein by immunohistochemistry. Both PAX3-FOXO1 and PAX7-FOXO1 were detected in HEK293 cells transfected with the corresponding cDNAs. Subsequently, we stained 26 primary tumor sections and a rhabdomyosarcoma tissue array and detected both fusion proteins with a positive predictive value of 100%, negative predictive value of 98%, specificity of 100% and a sensitivity of 91%. While tumors are stained homogenously in PAX3-FOXO1 cases, the staining pattern is heterogenous with scattered positive cells only in tumors expressing PAX7-FOXO1. No staining was observed in stromal cells, embryonal rhabdomyosarcoma, and fusion-negative rhabdomyosarcoma. These results demonstrate that mAbs specific for the chimeric oncoproteins PAX3-FOXO1 and PAX7-FOXO1 can be used efficiently for simple and fast subclassification of rhabdomyosarcoma in routine diagnostics via immunohistochemical detection.


Assuntos
Anticorpos Monoclonais/imunologia , Biomarcadores Tumorais/análise , Imuno-Histoquímica , Proteínas de Fusão Oncogênica/análise , Fatores de Transcrição Box Pareados/análise , Rabdomiossarcoma Alveolar/imunologia , Adolescente , Adulto , Animais , Especificidade de Anticorpos , Criança , Pré-Escolar , Feminino , Células HEK293 , Células HeLa , Humanos , Lactente , Masculino , Camundongos , Pessoa de Meia-Idade , Células NIH 3T3 , Proteínas de Fusão Oncogênica/imunologia , Fatores de Transcrição Box Pareados/imunologia , Valor Preditivo dos Testes , Reprodutibilidade dos Testes , Rabdomiossarcoma Alveolar/patologia , Adulto Jovem
5.
Hum Pathol ; 109: 37-44, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33301751

RESUMO

Salivary secretory carcinoma (SASC) is frequently associated with ETV6-neurotrophic tyrosine receptor kinase (NTRK) 3 fusion and more rarely with RET, MET, or ALK rearrangement. We aimed to elucidate the potential diagnostic utility of pan-tropomyosin receptor kinase (Trk) immunohistochemistry and its relationship with the fusion gene subtype in SASC. We examined 33 cases of SASC for immunoexpression of pan-Trk, ALK and ROS1, and gene rearrangement of the ETV6, NTRK3, and RET genes using fluorescence in situ hybridization (FISH) and reverse transcription-polymerase chain reaction (RT-PCR). Thirty (90.9%) of 33 SASCs harbored ETV6-NTRK3 fusion gene transcripts by RT-PCR and/or both ETV6 and NTRK3 gene rearrangements by FISH, and 3 cases (9.1%) had RET gene rearrangement. Most NTRK3-rearranged SASCs (27/33 cases; 81.8%) had conventional ETV6 exon 5-NTRK3 exon 15 fusion, whereas 2 cases (6.1%) had both the conventional fusion and a novel ETV6 exon 4-NTRK3 exon 15 fusion variant. In the remaining one case (3%), only FISH revealed both ETV6 and NTRK3 rearrangements, suggesting an ETV6-NTRK3 fusion with an as yet undetermined break point. All 30 SASCs with ETV6-NTRK3 fusion and/or NTRK3 rearrangement showed nuclear and cytoplasmic immunoreactivity for pan-Trk. In contrast, 3 SASCs with RET rearrangement showed negative or only weak cytoplasmic staining for pan-Trk. There was no case harboring ALK and ROS1 rearrangements. All 17 non-SASC tumors were negative for pan-Trk. The results suggest that nuclear and cytoplasmic immunoreactivity for pan-TRK may be helpful to identify ETV6-NTRK3-fused SASCs and to distinguish them from RET-rearranged SASCs and morphological mimics.


Assuntos
Neoplasias da Mama/imunologia , Carcinoma/imunologia , Proteínas de Fusão Oncogênica/imunologia , Proteínas Proto-Oncogênicas c-ret/imunologia , Adolescente , Adulto , Idoso , Biomarcadores Tumorais/análise , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Carcinoma/genética , Carcinoma/metabolismo , Criança , Feminino , Rearranjo Gênico/genética , Humanos , Imuno-Histoquímica/métodos , Pessoa de Meia-Idade , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/metabolismo , Proteínas Tirosina Quinases/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-ret/genética , Proteínas Proto-Oncogênicas c-ret/metabolismo , Receptores Proteína Tirosina Quinases/genética , Neoplasias das Glândulas Salivares/genética , Neoplasias das Glândulas Salivares/imunologia , Neoplasias das Glândulas Salivares/patologia , Tropomiosina/genética , Adulto Jovem
6.
Am J Surg Pathol ; 44(7): 922-933, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32141887

RESUMO

Synovial sarcoma (SS), an aggressive soft tissue sarcoma with a predilection for the extremities of young adults, harbors the pathognomonic t(X;18)(p11;q11) translocation, resulting in SS18-SSX rearrangements. SS includes monophasic, biphasic, and poorly differentiated variants, which show considerable histologic overlap with a range of other tumor types, making the diagnosis challenging on limited biopsies. Immunohistochemistry (IHC) is routinely used in the differential diagnosis; however, presently available markers lack specificity. Thus, cytogenetic or molecular genetic techniques are often employed to confirm the diagnosis. Here, we report the development and characterization of 2 novel antibodies: an SS18-SSX fusion-specific antibody (E9X9V, designed to the breakpoint) as well as an SSX-specific antibody (E5A2C, designed to the SSX C-terminus). We validated the selectivity and specificity of the antibodies using immunoblotting, immunoprecipitation, and chromatin immunoprecipitation followed by next-generation sequencing in SS cell lines and demonstrated that both antibodies capture SS18-SSX on chromatin at established target sites (eg, TLE1 and BCL2) genome-wide. Using IHC in whole sections from 400 tumors including 100 genetically confirmed cases of SS and 300 histologic mimics, the SS18-SSX fusion-specific antibody revealed strong diffuse nuclear staining in 95 of 100 (95%) SS cases, whereas none of the 300 control tumors showed any staining. The SSX antibody showed strong diffuse nuclear staining in all 100 (100%) SS cases; 13 (4%) of the 300 other tumors were also positive, 5 of which displayed >50% nuclear staining. In summary, a novel SS18-SSX fusion-specific antibody is highly sensitive (95%) and specific (100%) for SS, and an antibody to the SSX C-terminus is also highly sensitive (100%), but slightly less specific (96%). IHC using the SS18-SSX antibody could replace molecular genetic or cytogenetic testing in most cases, and these reagents together will also provide the research community with valuable tools for further biochemical and genomic interrogation of the SS18-SSX fusion protein.


Assuntos
Anticorpos Monoclonais , Biomarcadores Tumorais/imunologia , Proteínas de Neoplasias/imunologia , Proteínas de Fusão Oncogênica/imunologia , Proteínas Proto-Oncogênicas/imunologia , Proteínas Repressoras/imunologia , Sarcoma Sinovial/diagnóstico , Neoplasias de Tecidos Moles/diagnóstico , Animais , Biomarcadores Tumorais/genética , Estudos de Casos e Controles , Linhagem Celular Tumoral , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Immunoblotting , Imunoprecipitação , Hibridização in Situ Fluorescente , Proteínas de Neoplasias/genética , Proteínas de Fusão Oncogênica/genética , Proteínas Proto-Oncogênicas/genética , Coelhos , Proteínas Repressoras/genética , Sarcoma Sinovial/genética , Sensibilidade e Especificidade , Neoplasias de Tecidos Moles/genética
7.
Int J Mol Sci ; 20(15)2019 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-31370265

RESUMO

Osteosarcoma and Ewing sarcoma are the most common malignant primary bone tumors mainly occurring in children, adolescents and young adults. Current standard therapy includes multidrug chemotherapy and/or radiation specifically for Ewing sarcoma, associated with tumor resection. However, patient survival has not evolved for the past decade and remains closely related to the response of tumor cells to chemotherapy, reaching around 75% at 5 years for patients with localized forms of osteosarcoma or Ewing sarcoma but less than 30% in metastatic diseases and patients resistant to initial chemotherapy. Despite Ewing sarcoma being characterized by specific EWSR1-ETS gene fusions resulting in oncogenic transcription factors, currently, no targeted therapy could be implemented. It seems even more difficult to develop a targeted therapeutic strategy in osteosarcoma which is characterized by high complexity and heterogeneity in genomic alterations. Nevertheless, the common point between these different bone tumors is their ability to deregulate bone homeostasis and remodeling and divert them to their benefit. Therefore, targeting different actors of the bone tumor microenvironment has been hypothesized to develop new therapeutic strategies. In this context, it is well known that the Wnt/ß-catenin signaling pathway plays a key role in cancer development, including osteosarcoma and Ewing sarcoma as well as in bone remodeling. Moreover, recent studies highlight the implication of the Wnt/ß-catenin pathway in angiogenesis and immuno-surveillance, two key mechanisms involved in metastatic dissemination. This review focuses on the role played by this signaling pathway in the development of primary bone tumors and the modulation of their specific microenvironment.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Ósseas/tratamento farmacológico , Regulação Neoplásica da Expressão Gênica , Osteossarcoma/tratamento farmacológico , Sarcoma de Ewing/tratamento farmacológico , Microambiente Tumoral/efeitos dos fármacos , Adolescente , Neoplasias Ósseas/genética , Neoplasias Ósseas/imunologia , Neoplasias Ósseas/mortalidade , Osso e Ossos , Criança , Humanos , Metástase Linfática , Terapia de Alvo Molecular/métodos , Neovascularização Patológica/genética , Neovascularização Patológica/imunologia , Neovascularização Patológica/mortalidade , Neovascularização Patológica/prevenção & controle , Proteínas de Fusão Oncogênica/antagonistas & inibidores , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/imunologia , Osteossarcoma/genética , Osteossarcoma/imunologia , Osteossarcoma/mortalidade , Proteínas Proto-Oncogênicas c-ets/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-ets/genética , Proteínas Proto-Oncogênicas c-ets/imunologia , Proteína EWS de Ligação a RNA/antagonistas & inibidores , Proteína EWS de Ligação a RNA/genética , Proteína EWS de Ligação a RNA/imunologia , Sarcoma de Ewing/genética , Sarcoma de Ewing/imunologia , Sarcoma de Ewing/mortalidade , Análise de Sobrevida , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia , Via de Sinalização Wnt/efeitos dos fármacos , Adulto Jovem , beta Catenina/antagonistas & inibidores , beta Catenina/genética , beta Catenina/imunologia
8.
Sci Rep ; 9(1): 358, 2019 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-30674975

RESUMO

Osteosarcomas are characterized by highly disrupted genomes. Although osteosarcomas lack common fusions, we find evidence of many tumour specific gene-gene fusion transcripts, likely due to chromosomal rearrangements and expression of transcription-induced chimeras. Most of the fusions result in out-of-frame transcripts, potentially capable of producing long novel protein sequences and a plethora of neoantigens. To identify fusions, we explored RNA-sequencing data to obtain detailed knowledge of transcribed fusions, by creating a novel program to compare fusions identified by deFuse to de novo transcripts generated by Trinity. This allowed us to confirm the deFuse results and identify unusual splicing patterns associated with fusion events. Using various existing tools combined with this custom program, we developed a pipeline for the identification of fusion transcripts applicable as targets for immunotherapy. In addition to identifying candidate neoantigens associated with fusions, we were able to use the pipeline to establish a method for measuring the frequency of fusion events, which correlated to patient outcome, as well as highlight some similarities between canine and human osteosarcomas. The results of this study of osteosarcomas underscores the numerous benefits associated with conducting a thorough analysis of fusion events within cancer samples.


Assuntos
Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Neoplasias Ósseas/genética , Neoplasias Ósseas/imunologia , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/imunologia , Osteossarcoma/genética , Osteossarcoma/imunologia , Animais , Antiporters/genética , Neoplasias Ósseas/patologia , Linfócitos T CD8-Positivos/metabolismo , Proteínas CLOCK/genética , Proteínas de Transporte de Cátions/genética , Linhagem Celular Tumoral , Biologia Computacional/métodos , Epitopos/genética , Epitopos/imunologia , Perfilação da Expressão Gênica , Loci Gênicos , Instabilidade Genômica , Sequenciamento de Nucleotídeos em Larga Escala , Camundongos , Fases de Leitura Aberta , Osteossarcoma/patologia , Transcrição Gênica , Transcriptoma
9.
Am J Hematol ; 94(3): 338-345, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30575108

RESUMO

The prevalence, clinicopathologic correlates, and outcomes of previously untreated chronic lymphocytic leukemia (CLL) patients with IGH-BCL2 and IGH-BCL3 translocations are not well known. Using the Mayo Clinic CLL database, we identified patients seen between March 1, 2002 and September 30, 2016 who had FISH testing performed within 3 years of CLL diagnosis. The prognostic profile, time to first therapy (TTT), and overall survival (OS) of patients with IGH-BCL2 and IGH-BCL3 translocation were compared to patients without these abnormalities (non-IGH group). Of 1684 patients who met the inclusion criteria, 38 (2.2%) had IGH-BCL2, and 16 (0.9%) had IGH-BCL3 translocation at diagnosis. Patients with IGH-BCL3 translocation were more likely to have high and very-high CLL-International Prognostic Index, compared to patients with IGH-BCL2 translocation and the non-IGH group. The 5-year probability of requiring therapy was significantly higher for IGH-BCL3 compared to IGH-BCL2 and non-IGH groups (84% vs 33% vs 29%, respectively, P < 0.0001). The 5-year OS was significantly shorter for IGH-BCL3 compared to IGH-BCL2 and non-IGH groups (45% vs 89% vs 86%, respectively, P < 0.0001). On multivariable analyses, IGH-BCL3 translocation was associated with a shorter TTT (hazard ratio [HR] = 2.7; P = 0.005) and shorter OS (HR = 5.5; P < 0.0001); IGH-BCL2 translocation did not impact TTT and OS. In conclusion, approximately 3% of all newly diagnosed CLL patients have either an IGH-BCL2 or IGH-BCL3 translocation. Patients with IGH-BCL3 translocations have a distinct prognostic profile and outcome. These results support the inclusion of an IGH probe during the routine evaluation of FISH abnormalities in newly diagnosed CLL.


Assuntos
Proteína 3 do Linfoma de Células B/genética , Cadeias Pesadas de Imunoglobulinas/genética , Leucemia Linfocítica Crônica de Células B/genética , Proteínas de Fusão Oncogênica/genética , Proteínas Proto-Oncogênicas c-bcl-2/genética , Adenina/análogos & derivados , Adulto , Idoso , Idoso de 80 Anos ou mais , Antraciclinas/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Antineoplásicos Imunológicos/uso terapêutico , Proteína 3 do Linfoma de Células B/imunologia , Estudos de Coortes , Feminino , Expressão Gênica , Humanos , Cadeias Pesadas de Imunoglobulinas/imunologia , Leucemia Linfocítica Crônica de Células B/diagnóstico , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico , Leucemia Linfocítica Crônica de Células B/mortalidade , Masculino , Pessoa de Meia-Idade , Proteínas de Fusão Oncogênica/imunologia , Piperidinas , Prognóstico , Proteínas Proto-Oncogênicas c-bcl-2/imunologia , Pirazóis/uso terapêutico , Pirimidinas/uso terapêutico , Análise de Sobrevida , Translocação Genética , Resultado do Tratamento
10.
Am J Med Sci ; 356(1): 72-78, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-30049331

RESUMO

BACKGROUND: This study was designed to examine the effect of transmembrane protease serine 2 ETS-related gene (TMPRSS2-ERG) fusion on regulatory T cells and tumor growth in prostate cancer, which may provide a new potential therapeutic direction for PCa. METHODS: The effect of TMPRSS2-ERG fusion on the migration of Treg cells and tumor growth in a mouse model was investigated in vitro and in vivo. TMPRSS2-ERG fusion in biopsy tissues was performed by fluorescence in situ hybridization and the expression of ERG and Forkhead box P3 was detected by gel electrophoresis, real-time quantitative reverse transcription polymerase chain reaction and Western blot. Enzyme-linked immunosorbent assay and flow cytometry were used to analyze transforming growth factor ß levels and the number of regulatory T cells, respectively. Finally, the infiltration of regulatory T cells was analyzed by Forkhead box P3 immunohistochemistry. RESULTS: Fluorescence in situ hybridization analysis showed that the TMPRSS2-ERG fusion gene was positive in prostate cancer and that the messenger RNA and protein expression of ERG were significantly up-regulated in prostate cancer biopsy tissues. Furthermore, the number of regulatory T cells and the levels of Forkhead box P3 and transforming growth factor ß were significantly increased in prostate cancer. TMPRSS2-ERG fusion increased the migration and activation of regulatory T cells in vitro and promoted subcutaneous tumor size and regulatory T cells infiltration in mouse models. CONCLUSIONS: TMPRSS2-ERG fusion can regulate the recruitment and infiltration of regulatory T cells to promote tumor growth in prostate cancer.


Assuntos
Proteínas de Fusão Oncogênica/imunologia , Neoplasias da Próstata/imunologia , Linfócitos T Reguladores/imunologia , Animais , Fatores de Transcrição Forkhead/biossíntese , Fatores de Transcrição Forkhead/imunologia , Xenoenxertos , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Transplante de Neoplasias , Proteínas de Fusão Oncogênica/biossíntese , Células PC-3 , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Linfócitos T Reguladores/patologia , Análise Serial de Tecidos
11.
Curr Pharm Biotechnol ; 19(1): 5-18, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29667553

RESUMO

BACKGROUND: Cancer is one of the leading causes of death worldwide. Over the years, a number of conventional cytotoxic approaches for neoplastic diseases has been developed. However, due to their limited effectiveness in accordance with the heterogeneity of cancer cells, there is a constant search for therapeutic approaches with improved outcome, such as immunotherapy that utilizes and enhances the normal capacity of the patient's immune system. METHODS: Chimeric Antigen Receptor (CAR) T-cell therapy involves genetic modification of patient's autologous T-cells to express a CAR specific for a tumor antigen, following by ex vivo cell expansion and re-infusion back to the patient. CARs are fusion proteins of a selected single-chain fragment variable from a specific monoclonal antibody and one or more T-cell receptor intracellular signaling domains. This T-cell genetic modification may occur either via viral-based gene transfer methods or nonviral methods, such as DNA-based transposons, CRISPR/Cas9 technology or direct transfer of in vitro transcribed-mRNA by electroporation. RESULTS: Clinical trials have shown very promising results in end-stage patients with a full recovery of up to 92% in Acute Lymphocytic Leukemia. Despite such results in hematological cancers, the effective translation of CAR T-cell therapy to solid tumors and the corresponding clinical experience is limited due to therapeutic barriers, like CAR T-cell expansion, persistence, trafficking, and fate within tumors. CONCLUSION: In this review, the basic design of CARs, the main genetic modification strategies, the safety matters as well as the initial clinical experience with CAR T-cells are described.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/tendências , Imunoterapia/tendências , Neoplasias/imunologia , Neoplasias/terapia , Linfócitos T/imunologia , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Antígenos de Neoplasias/uso terapêutico , Terapia Baseada em Transplante de Células e Tecidos/métodos , Técnicas de Transferência de Genes/tendências , Humanos , Imunoterapia/métodos , Neoplasias/genética , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/imunologia , Proteínas de Fusão Oncogênica/uso terapêutico , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/uso terapêutico , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/uso terapêutico
12.
Manag Care ; 26(10): 28-30, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-29068298

RESUMO

FDA approval of the CAR T-cell therapy for leukemia could usher in an era of genetically engineered, individually tailored immunotherapies. But tap those brakes. Long-term results are in short supply-and there's that $475,000 price tag. Or is it a $750,000 price tag?


Assuntos
Antineoplásicos/economia , Indústria Farmacêutica/economia , Leucemia de Células B/tratamento farmacológico , Receptores de Antígenos de Linfócitos T , Custos de Medicamentos , Humanos , Leucemia de Células B/imunologia , Proteínas de Fusão Oncogênica/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/imunologia , Estados Unidos
13.
Clin Cancer Res ; 23(24): 7596-7607, 2017 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-28954787

RESUMO

Purpose: Gene fusions are frequently found in prostate cancer and may result in the formation of unique chimeric amino acid sequences (CASQ) that span the breakpoint of two fused gene products. This study evaluated the potential for fusion-derived CASQs to be a source of tumor neoepitopes, and determined their relationship to patterns of immune signatures in prostate cancer patients.Experimental Design: A computational strategy was used to identify CASQs and their corresponding predicted MHC class I epitopes using RNA-Seq data from The Cancer Genome Atlas of prostate tumors. In vitro peptide-specific T-cell expansion was performed to identify CASQ-reactive T cells. A multivariate analysis was used to relate patterns of in silico-predicted tumor-infiltrating immune cells with prostate tumors harboring these mutational events.Results: Eighty-seven percent of tumors contained gene fusions with a mean of 12 per tumor. In total, 41% of fusion-positive tumors were found to encode CASQs. Within these tumors, 87% gave rise to predicted MHC class I-binding epitopes. This observation was more prominent when patients were stratified into low- and intermediate/high-risk categories. One of the identified CASQ from the recurrent TMPRSS2:ERG type VI fusion contained several high-affinity HLA-restricted epitopes. These peptides bound HLA-A*02:01 in vitro and were recognized by CD8+ T cells. Finally, the presence of fusions and CASQs were associated with expression of immune cell infiltration.Conclusions: Mutanome analysis of gene fusion-derived CASQs can give rise to patient-specific predicted neoepitopes. Moreover, these fusions predicted patterns of immune cell infiltration within a subgroup of prostate cancer patients. Clin Cancer Res; 23(24); 7596-607. ©2017 AACR.


Assuntos
Epitopos de Linfócito T/genética , Antígeno HLA-A2/genética , Proteínas de Fusão Oncogênica/genética , Neoplasias da Próstata/imunologia , Sequência de Aminoácidos/genética , Linfócitos T CD8-Positivos/imunologia , Análise Mutacional de DNA , Epitopos de Linfócito T/imunologia , Genes MHC Classe I/genética , Genes MHC Classe I/imunologia , Antígeno HLA-A2/imunologia , Humanos , Masculino , Proteínas de Fusão Oncogênica/imunologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Ligação Proteica , Linfócitos T/imunologia
14.
BMC Immunol ; 18(1): 40, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28810829

RESUMO

BACKGROUND: Cancer therapeutic vaccine induced cytotoxic T cell (CTL) responses are pivotal for the killing of tumour cells. Blocking interleukin 10 (IL-10) signalling at the time of immunization increases vaccine induced CTL responses and improves prevention of tumour growth in animal models compared to immunization without an IL-10 signalling blockade. Therefore, this immunization strategy may have potential to curtail cancer in a clinical setting. However, IL-10 deficiency leads to autoimmune disease in the gut. Blocking IL-10 at the time of immunization may result in unwanted side effects, especially immune-pathological diseases in the intestine. METHODS: We investigated whether blocking IL-10 at the time of immunization results in intestinal inflammation responses in a mouse TC-1 tumour model and in a NOD autoimmune disease prone mouse model. RESULTS: We now show that blocking IL-10 at the time of immunization increases IL-10 production by CD4+ T cells in the spleen and draining lymph nodes, and does not result in blood cell infiltration to the intestines leading to intestinal pathological changes. Moreover, immunization with papillomavirus like particles combined with simultaneously blocking IL-10 signalling does not increase the incidence of autoimmune disease in Non-obese diabetic (NOD) mice. CONCLUSIONS: Our results indicate that immunization with an IL-10 inhibitor may facilitate the generation of safe, effective therapeutic vaccines against chronic viral infection and cancer.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Imunização/efeitos adversos , Imunização/métodos , Interleucina-10/antagonistas & inibidores , Intestinos/imunologia , Transdução de Sinais/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Anticorpos/imunologia , Doenças Autoimunes/imunologia , Linfócitos T CD4-Positivos/metabolismo , Proliferação de Células , Modelos Animais de Doenças , Feminino , Interleucina-10/imunologia , Interleucina-10/metabolismo , Intestinos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD/imunologia , Camundongos Knockout , Proteínas de Fusão Oncogênica/imunologia , Proteínas Oncogênicas Virais/imunologia , Proteínas E7 de Papillomavirus/química , Proteínas E7 de Papillomavirus/imunologia , Baço/citologia , Baço/imunologia
15.
Cancer Discov ; 7(9): 924, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28751490

RESUMO

An expert panel recommended approval of Novartis's experimental chimeric antigen T-cell therapy, tisagenlecleucel, for children and young adults with relapsed or refractory B-cell acute lymphoblastic leukemia. The therapy would be the first of its kind approved for cancer and has the potential to transform standard of care for advanced blood cancers.


Assuntos
Leucemia/tratamento farmacológico , Proteínas de Fusão Oncogênica/uso terapêutico , Receptores de Antígenos de Linfócitos T/uso terapêutico , Adolescente , Criança , Pré-Escolar , Feminino , Humanos , Leucemia/imunologia , Masculino , Proteínas de Fusão Oncogênica/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/imunologia , Adulto Jovem
16.
J Immunother Cancer ; 5: 47, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28649380

RESUMO

BACKGROUND: The expansion of antigen-specific CD8 T cells is important in generating an effective and long-lasting immune response to tumors and viruses. Glucocorticoid-induced tumor necrosis factor receptor family-related receptor (GITR) is a co-stimulatory receptor that binds the GITR ligand (GITRL). Agonism of GITR can produce important signals that drive expansion of effector T cell populations. METHODS: We explored two separate murine tumor models, CT26 and TC-1, for responsiveness to GITR Ligand Fusion Protein(GITRL-FP) monotherapy. In TC-1, GITRL-FP was also combined with concurrent administration of an E7-SLP vaccine. We evaluated tumor growth inhibition by tumor volume measurements as well as changes in CD8 T cell populations and function including cytokine production using flow cytometry. Additionally, we interrogated how these therapies resulted in tumor antigen-specific responses using MHC-I dextramer staining and antigen-specific restimulations. RESULTS: In this study, we demonstrate that a GITR ligand fusion protein (GITRL-FP) is an effective modulator of antigen-specific CD8 T cells. In a CT26 mouse tumor model, GITRL-FP promoted expansion of antigen-specific T cells, depletion of regulatory T cells (Tregs), and generation of long-lasting CD8 T cell memory. This memory expansion was dependent on the dose of GITRL-FP and resulted in complete tumor clearance and protection from tumor rechallenge. In contrast, in TC-1 tumor-bearing mice, GITRL-FP monotherapy could not prime an antigen-specific CD8 T cell response and was unable to deplete Tregs. However, when combined with a vaccine targeting E7, treatment with GITRL-FP resulted in an augmentation of the vaccine-induced antigen-specific CD8 T cells, the depletion of Tregs, and a potent antitumor immune response. In both model systems, GITR levels on antigen-specific CD8 T cells were higher than on all other CD8 T cells, and GITRL-FP interacted directly with primed antigen-specific CD8 T cells. CONCLUSIONS: When taken together, our results demonstrate that the delivery of GITRL-FP as a therapeutic can promote anti-tumor responses in the presence of tumor-specific CD8 T cells. These findings support further study into combination partners for GITRL-FP that may augment CD8 T-cell priming as well as provide hypotheses that can be tested in human clinical trials exploring GITR agonists including GITRL-FP.


Assuntos
Antígenos de Neoplasias/imunologia , Neoplasias do Colo/tratamento farmacológico , Proteína Relacionada a TNFR Induzida por Glucocorticoide/genética , Proteínas de Fusão Oncogênica/genética , Animais , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/imunologia , Neoplasias do Colo/genética , Neoplasias do Colo/imunologia , Neoplasias do Colo/patologia , Modelos Animais de Doenças , Feminino , Proteína Relacionada a TNFR Induzida por Glucocorticoide/agonistas , Proteína Relacionada a TNFR Induzida por Glucocorticoide/imunologia , Humanos , Camundongos , Proteínas de Fusão Oncogênica/antagonistas & inibidores , Proteínas de Fusão Oncogênica/imunologia , Receptores do Fator de Necrose Tumoral/genética , Receptores do Fator de Necrose Tumoral/imunologia , Transdução de Sinais/efeitos dos fármacos , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Fatores de Necrose Tumoral/genética , Fatores de Necrose Tumoral/imunologia
17.
Mol Biol (Mosk) ; 51(2): 274-287, 2017.
Artigo em Russo | MEDLINE | ID: mdl-28537234

RESUMO

Early results from clinical trials of autologous chimeric antigen receptor (CAR)-expressing T cells for the therapy of B-cell malignancies have encouraged extending the potency of this therapy to other cancers. However, the success of using CAR T-cells to treat patients with solid tumors has been limited. In this review, we summarize current knowledge on the design and applications of CARs for the targeted therapy of cancer. We describe existing issues that limit the widespread application of CAR T cells and discuss the optimization steps needed to further improve safety and efficacy of this therapeutic platform.


Assuntos
Neoplasias Hematológicas , Imunoterapia/métodos , Proteínas de Fusão Oncogênica , Receptores de Antígenos de Linfócitos T , Linfócitos T , Animais , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/imunologia , Neoplasias Hematológicas/terapia , Humanos , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/imunologia , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/imunologia , Linfócitos T/transplante
18.
Cancer Discov ; 7(4): 342, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28193774

RESUMO

Two infants with relapsed, refractory B-cell acute lymphoblastic leukemia went into complete remission after being treated with CD19-targeting CAR T cells derived from an unmatched donor. The study is the first to demonstrate that a universal form of CAR T-cell therapy can be safely utilized.


Assuntos
Antígenos CD19/imunologia , Imunoterapia , Leucemia de Células B/terapia , Receptores de Antígenos de Linfócitos T/uso terapêutico , Linfócitos T/imunologia , Antígenos CD19/genética , Terapia Baseada em Transplante de Células e Tecidos , Humanos , Lactente , Leucemia de Células B/genética , Leucemia de Células B/imunologia , Leucemia de Células B/patologia , Proteínas de Fusão Oncogênica/imunologia , Proteínas de Fusão Oncogênica/uso terapêutico , Receptores de Antígenos de Linfócitos T/imunologia , Indução de Remissão
19.
Clin Cancer Res ; 23(14): 3499-3509, 2017 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-28183713

RESUMO

Purpose: The multiple mechanisms used by solid tumors to suppress tumor-specific immune responses are a major barrier to the success of adoptively transferred tumor-specific T cells. As viruses induce potent innate and adaptive immune responses, we hypothesized that the immunogenicity of viruses could be harnessed for the treatment of solid tumors if virus-specific T cells (VST) were modified with tumor-specific chimeric antigen receptors (CAR). We tested this hypothesis using VZV-specific T cells (VZVST) expressing a CAR for GD2, a disialoganglioside expressed on neuroblastoma and certain other tumors, so that the live-attenuated VZV vaccine could be used for in vivo stimulation.Experimental Design: We generated GMP-compliant, GD2.CAR-modified VZVSTs from healthy donors and cancer patients by stimulation of peripheral blood mononuclear cells with overlapping peptide libraries spanning selected VZV antigens, then tested their ability to recognize and kill GD2- and VZV antigen-expressing target cells.Results: Our choice of VZV antigens was validated by the observation that T cells specific for these antigens expanded in vivo after VZV vaccination. VZVSTs secreted cytokines in response to VZV antigens, killed VZV-infected target cells and limited infectious virus spread in autologous fibroblasts. However, while GD2.CAR-modified VZVSTs killed neuroblastoma cell lines on their first encounter, they failed to control tumor cells in subsequent cocultures. Despite this CAR-specific dysfunction, CAR-VZVSTs retained functional specificity for VZV antigens via their TCRs and GD2.CAR function was partially rescued by stimulation through the TCR or exposure to dendritic cell supernatants.Conclusions: Vaccination via the TCR may provide a means to reactivate CAR-T cells rendered dysfunctional by the tumor microenvironment (NCT01953900). Clin Cancer Res; 23(14); 3499-509. ©2017 AACR.


Assuntos
Neuroblastoma/tratamento farmacológico , Proteínas de Fusão Oncogênica/administração & dosagem , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T Citotóxicos/imunologia , Adolescente , Adulto , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Criança , Pré-Escolar , Citotoxicidade Imunológica/imunologia , Células Dendríticas/imunologia , Feminino , Humanos , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Masculino , Camundongos , Pessoa de Meia-Idade , Neuroblastoma/imunologia , Neuroblastoma/patologia , Proteínas de Fusão Oncogênica/imunologia , Receptores de Antígenos de Linfócitos T/antagonistas & inibidores , Microambiente Tumoral/imunologia , Vacinação , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
20.
Clin Cancer Res ; 23(13): 3416-3427, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28069723

RESUMO

Purpose: To generate and characterize a murine GITR ligand fusion protein (mGITRL-FP) designed to maximize valency and the potential to agonize the GITR receptor for cancer immunotherapy.Experimental Design: The EC50 value of the mGITRL-FP was compared with an anti-GITR antibody in an in vitro agonistic cell-based reporter assay. We assessed the impact of dose, schedule, and Fc isotype on antitumor activity and T-cell modulation in the CT26 tumor model. The activity of the mGITRL-FP was compared with an agonistic murine OX40L-FP targeting OX40, in CT26 and B16F10-Luc2 tumor models. Combination of the mGITRL-FP with antibodies targeting PD-L1, PD-1, or CTLA-4 was analyzed in mice bearing CT26 tumors.Results: The mGITRL-FP had an almost 50-fold higher EC50 value compared with an anti-murine GITR antibody. Treatment of CT26 tumor-bearing mice with mGITRL-FP-mediated significant antitumor activity that was dependent on isotype, dose, and duration of exposure. The antitumor activity could be correlated with the increased proliferation of peripheral CD8+ and CD4+ T cells and a significant decrease in the frequency of intratumoral Tregs. The combination of mGITRL-FP with mOX40L-FP or checkpoint inhibitor antagonists enhanced antitumor immunity above that of monotherapy treatment.Conclusions: These results suggest that therapeutically targeting GITR represents a unique approach to cancer immunotherapy and suggests that a multimeric fusion protein may provide increased agonistic potential versus an antibody. In addition, these data provide, for the first time, early proof of concept for the potential combination of GITR targeting agents with OX40 agonists and PD-L1 antagonists. Clin Cancer Res; 23(13); 3416-27. ©2017 AACR.


Assuntos
Proteína Relacionada a TNFR Induzida por Glucocorticoide/imunologia , Melanoma Experimental/imunologia , Proteínas de Fusão Oncogênica/administração & dosagem , Fatores de Necrose Tumoral/imunologia , Animais , Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/imunologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Antígeno CTLA-4/antagonistas & inibidores , Antígeno CTLA-4/imunologia , Modelos Animais de Doenças , Proteína Relacionada a TNFR Induzida por Glucocorticoide/administração & dosagem , Humanos , Melanoma Experimental/genética , Melanoma Experimental/terapia , Glicoproteínas de Membrana/agonistas , Glicoproteínas de Membrana/imunologia , Camundongos , Ligante OX40 , Proteínas de Fusão Oncogênica/imunologia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/imunologia , Fatores de Necrose Tumoral/agonistas , Fatores de Necrose Tumoral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA