Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Immunology ; 148(3): 276-86, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27012310

RESUMO

Interleukin-2 (IL-2) is a critical regulator of immune homeostasis through its non-redundant role in regulatory T (Treg) cell biology. There is major interest in therapeutic modulation of the IL-2 pathway to promote immune activation in the context of tumour immunotherapy or to enhance immune suppression in the context of transplantation, autoimmunity and inflammatory diseases. Antibody-mediated targeting of the high-affinity IL-2 receptor α chain (IL-2Rα or CD25) offers a direct mechanism to target IL-2 biology and is being actively explored in the clinic. In mouse models, the rat anti-mouse CD25 clone PC61 has been used extensively to investigate the biology of IL-2 and Treg cells; however, there has been controversy and conflicting data on the exact in vivo mechanistic function of PC61. Engineering antibodies to alter Fc/Fc receptor interactions can significantly alter their in vivo function. In this study, we re-engineered the heavy chain constant region of an anti-CD25 monoclonal antibody to generate variants with highly divergent Fc effector function. Using these anti-CD25 Fc variants in multiple mouse models, we investigated the in vivo impact of CD25 blockade versus depletion of CD25(+) Treg cells on immune homeostasis. We report that immune homeostasis can be maintained during CD25 blockade but aberrant T-cell activation prevails when CD25(+) Treg cells are actively depleted. These results clarify the impact of PC61 on Treg cell biology and reveal an important distinction between CD25 blockade and depletion of CD25(+) Treg cells. These findings should inform therapeutic manipulation of the IL-2 pathway by targeting the high-affinity IL-2R.


Assuntos
Anticorpos Monoclonais/farmacologia , Imunoglobulina G/farmacologia , Imunoterapia , Interleucina-2/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Linfócitos T Reguladores/efeitos dos fármacos , Animais , Anticorpos Monoclonais/genética , Anticorpos Antivirais/imunologia , Autoimunidade/imunologia , Fatores de Transcrição Forkhead/metabolismo , Homeostase/efeitos dos fármacos , Imunoglobulina G/genética , Terapia de Imunossupressão , Interleucina-2/imunologia , Depleção Linfocítica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Engenharia de Proteínas , Ratos , Receptores de IgG/genética , Proteínas Recombinantes de Fusão/genética , Linfócitos T Reguladores/imunologia
2.
Immunology ; 147(4): 399-413, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26694902

RESUMO

Retinoic acid receptor-related orphan nuclear receptor γ (RORγ) orchestrates a pro-inflammatory gene expression programme in multiple lymphocyte lineages including T helper type 17 (Th17) cells, γδ T cells, innate lymphoid cells and lymphoid tissue inducer cells. There is compelling evidence that RORγ-expressing cells are relevant targets for therapeutic intervention in the treatment of autoimmune and inflammatory diseases. Unlike Th17 cells, where RORγ expression is induced under specific pro-inflammatory conditions, γδ T cells and other innate-like immune cells express RORγ in the steady state. Small molecule mediated disruption of RORγ function in cells with pre-existing RORγ transcriptional complexes represents a significant and challenging pharmacological hurdle. We present data demonstrating that a novel, selective and potent small molecule RORγ inhibitor can block the RORγ-dependent gene expression programme in both Th17 cells and RORγ-expressing γδ T cells as well as a disease-relevant subset of human RORγ-expressing memory T cells. Importantly, systemic administration of this inhibitor in vivo limits pathology in an innate lymphocyte-driven mouse model of psoriasis.


Assuntos
Doenças Autoimunes/etiologia , Doenças Autoimunes/metabolismo , Benzamidas/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Piridinas/farmacologia , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/imunologia , Dermatite/tratamento farmacológico , Dermatite/imunologia , Dermatite/metabolismo , Dermatite/patologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Humanos , Memória Imunológica/efeitos dos fármacos , Interleucina-17/metabolismo , Camundongos , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/antagonistas & inibidores , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Linfócitos T Auxiliares-Indutores/citologia , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/metabolismo , Células Th17/citologia , Células Th17/imunologia , Células Th17/metabolismo
3.
Bioorg Med Chem Lett ; 25(15): 2991-7, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-26048806

RESUMO

RORγt is a pivotal regulator of a pro-inflammatory gene expression program implicated in the pathology of several major human immune-mediated diseases. Evidence from mouse models demonstrates that genetic or pharmacological inhibition of RORγ activity can block the production of pathogenic cytokines, including IL-17, and convey therapeutic benefit. We have identified and developed a biaryl-carboxylamide series of RORγ inverse agonists via a structure based design approach. Co-crystal structures of compounds 16 and 48 supported the design approach and confirmed the key interactions with RORγ protein; the hydrogen bonding with His479 was key to the significant improvement in inverse agonist effect. The results have shown this is a class of potent and selective RORγ inverse agonists, with demonstrated oral bioavailability in rodents.


Assuntos
Amidas/química , Amidas/farmacologia , Compostos de Bifenilo/química , Compostos de Bifenilo/farmacologia , Agonismo Inverso de Drogas , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/agonistas , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/antagonistas & inibidores , Amidas/farmacocinética , Animais , Compostos de Bifenilo/farmacocinética , Linhagem Celular , Citocinas/imunologia , Descoberta de Drogas , Humanos , Ligação de Hidrogênio , Interleucina-17/imunologia , Camundongos , Simulação de Acoplamento Molecular , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/imunologia , Ratos
4.
Cell Chem Biol ; 28(6): 802-812.e6, 2021 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-33333026

RESUMO

The recent development of successful CAR (chimeric antigen receptor) T cell therapies has been accompanied by a need to better control potentially fatal toxicities that can arise from adverse immune reactions. Here we present a ligand-controlled CAR system, based on the IKZF3 ZF2 ß-hairpin IMiD-inducible degron, which allows for the reversible control of expression levels of type I membrane proteins, including CARs. Testing this system in an established mouse xenotransplantation model for acute lymphoblastic leukemia, we validate the ability of the CAR19-degron to target and kill CD19-positive cells displaying complete control/clearance of the tumor. We also demonstrate that the activity of CAR19-degron can be regulated in vivo when dosing a US Food and Drug Administration-approved drug, lenalidomide.


Assuntos
Fator de Transcrição Ikaros/imunologia , Fatores Imunológicos/farmacologia , Receptores de Antígenos Quiméricos/imunologia , Linfócitos T/imunologia , Adolescente , Animais , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Feminino , Humanos , Fator de Transcrição Ikaros/química , Fatores Imunológicos/química , Masculino , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos NOD , Camundongos SCID , Pessoa de Meia-Idade , Estrutura Molecular , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/patologia , Receptores de Antígenos Quiméricos/genética , Adulto Jovem
5.
Nat Med ; 27(5): 842-850, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33888899

RESUMO

While CD19-directed chimeric antigen receptor (CAR) T cells can induce remission in patients with B cell acute lymphoblastic leukemia (ALL), a large subset relapse with CD19- disease. Like CD19, CD22 is broadly expressed by B-lineage cells and thus serves as an alternative immunotherapy target in ALL. Here we present the composite outcomes of two pilot clinical trials ( NCT02588456 and NCT02650414 ) of T cells bearing a 4-1BB-based, CD22-targeting CAR in patients with relapsed or refractory ALL. The primary end point of these studies was to assess safety, and the secondary end point was antileukemic efficacy. We observed unexpectedly low response rates, prompting us to perform detailed interrogation of the responsible CAR biology. We found that shortening of the amino acid linker connecting the variable heavy and light chains of the CAR antigen-binding domain drove receptor homodimerization and antigen-independent signaling. In contrast to CD28-based CARs, autonomously signaling 4-1BB-based CARs demonstrated enhanced immune synapse formation, activation of pro-inflammatory genes and superior effector function. We validated this association between autonomous signaling and enhanced function in several CAR constructs and, on the basis of these observations, designed a new short-linker CD22 single-chain variable fragment for clinical evaluation. Our findings both suggest that tonic 4-1BB-based signaling is beneficial to CAR function and demonstrate the utility of bedside-to-bench-to-bedside translation in the design and implementation of CAR T cell therapies.


Assuntos
Ligante 4-1BB/metabolismo , Imunoterapia Adotiva/métodos , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Receptores de Antígenos Quiméricos/metabolismo , Lectina 2 Semelhante a Ig de Ligação ao Ácido Siálico/metabolismo , Linfócitos T/transplante , Adulto , Animais , Antígenos CD19/metabolismo , Linfócitos B/imunologia , Antígenos CD28/genética , Células Cultivadas , Criança , Pré-Escolar , Feminino , Humanos , Masculino , Camundongos , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Linfócitos T/imunologia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Cancer Res ; 67(16): 7875-84, 2007 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-17699794

RESUMO

Antiangiogenic immunotherapy, which targets molecules critical to tumor angiogenesis, is expected to counteract the negative effect of tumor cell genetic instability on the outcome of immunotherapy targeting tumor antigens. Previously, targeting of individual angiogenic molecules has been shown to inhibit tumor angiogenesis and limit tumor growth. Nevertheless, this approach may be bypassed by redundant angiogenic pathways. To overcome this limitation, we have developed an immunization strategy targeting multiple molecules critical to angiogenesis. To this end, hybrids of dendritic cells (DC) and syngeneic endothelial cells (EC) were used as immunogens, because (a) whole EC express multiple molecules involved in angiogenesis and (b) DC tumor cell hybrids are effective in generating self-antigen-specific immune responses. The immunization strategy included the administration of an agonist 4-1BB-specific monoclonal antibody (mAb), because it augments self-antigen-specific immune responses elicited by DC hybrids. Immunization of mice with DC-EC hybrids and 4-1BB-specific mAb inhibited the growth of B16.F10 melanoma and MC38 colon adenocarcinoma tumors. This effect is mediated by EC-specific CD4+ and CD8+ T-cell responses, which markedly inhibited tumor angiogenesis. No therapy-related side effects, except minor and transient hematologic changes, were observed. Our findings represent a useful background for the design of antiangiogenic immunotherapeutic strategies to control tumor growth in a clinical setting.


Assuntos
Adenocarcinoma/irrigação sanguínea , Neoplasias do Colo/irrigação sanguínea , Células Dendríticas/imunologia , Células Endoteliais/imunologia , Imunoterapia Adotiva/métodos , Melanoma Experimental/irrigação sanguínea , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia , Adenocarcinoma/imunologia , Adenocarcinoma/terapia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Especificidade de Anticorpos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Neoplasias do Colo/imunologia , Neoplasias do Colo/terapia , Feminino , Células Híbridas , Melanoma Experimental/imunologia , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Patológica/imunologia , Neovascularização Patológica/terapia
7.
Cancer Res ; 65(15): 6976-83, 2005 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-16061683

RESUMO

To overcome unresponsiveness to the self-high molecular weight melanoma-associated antigen (HMW-MAA) in hosts with constitutive HMW-MAA expression, we have used as immunogen the anti-idiotypic monoclonal antibody (mAb) MK2-23, which mimics the antigenic determinant recognized by the anti-HMW-MAA mAb 763.74. In a phase I/II clinical trial, anti-idiotypic mAb MK2-23, conjugated to keyhole limpet hemocyanin (KLH) as a carrier and given with Bacillus Calmette-Guerin (BCG) as an adjuvant, elicited HMW-MAA-specific antibodies in about 60% of the immunized melanoma patients. The immune response was associated with survival prolongation. However, safety and standardization issues associated with the use of KLH and BCG in the clinical setting have prompted us to develop alternative immunization strategies. Conjugation of human interleukin 2 (IL-2) to mAb MK2-23 variable regions covalently linked to human immunoglobulin constant regions enhanced mAb MK2-23 immunogenicity in BALB/c mice to an extent similar to that induced by mAb MK2-23 conjugated to KLH and given with Freund's adjuvant. As determined by the level of serum antibodies and delayed-type hypersensitivity responses to HMW-MAA-bearing melanoma cells, immunization of mice with the MK2-23-IL-2 fusion protein elicited more robust humoral and cellular responses, respectively, than immunization with KLH-conjugated mAb MK2-23 and separate administration of IL-2. The immunogenicity of the fusion protein is dependent on IL-2 conjugation, because immunization of mice with either mAb MK2-23 or chimeric mAb MK2-23, in combination with IL-2, was not as effective in eliciting HMW-MAA-specific immune responses. These results suggest that the MK2-23-IL-2 fusion protein represents a useful immunogen to implement active specific immunotherapy in patients with melanoma, because it bypasses the requirement for KLH conjugation and adjuvant administration.


Assuntos
Anticorpos Anti-Idiotípicos/imunologia , Antígenos de Neoplasias/imunologia , Imunotoxinas/imunologia , Interleucina-2/imunologia , Animais , Anticorpos Monoclonais/imunologia , Linhagem Celular Tumoral , Hemocianinas/imunologia , Humanos , Linfócitos/imunologia , Melanoma/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Mimetismo Molecular/imunologia , Proteínas Recombinantes de Fusão/imunologia
8.
J Interferon Cytokine Res ; 37(1): 20-31, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27835061

RESUMO

Because of its tumor-suppressive effect, interferon-based therapy has been used for the treatment of melanoma. However, limited data are available regarding the antitumor effects of pegylated interferons, either alone or in combination with approved anticancer drugs. We report that treatment of human WM-266-4 melanoma cells with peginterferon beta-1a induced apoptotic markers. Additionally, peginterferon beta-1a significantly inhibited the growth of human SK-MEL-1, A-375, and WM-266-4 melanoma xenografts established in immunocompromised mice. Peginterferon beta-1a regressed large, established WM-266-4 xenografts in nude mice. Treatment of SK-MEL-1 tumor-bearing mice with a combination of peginterferon beta-1a and the MEK inhibitor PD325901 ((R)-N-(2,3-dihydroxypropoxy)-3,4-difluoro-2-(2-fluoro-4-iodophenylamino)benzamide) significantly improved tumor growth inhibition compared with either agent alone. Examination of the antitumor activity of peginterferon beta-1a in combination with approved anticancer drugs in breast and renal carcinomas revealed improved antitumor activity in these preclinical xenograft models, as did the combination of peginterferon beta-1a and bevacizumab in a colon carcinoma xenograft model.


Assuntos
Antineoplásicos/farmacologia , Interferon beta/farmacologia , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Polietilenoglicóis/farmacologia , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacocinética , Antineoplásicos Imunológicos/farmacologia , Apoptose/efeitos dos fármacos , Biomarcadores Tumorais , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Sobrevivência Celular , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/patologia , Modelos Animais de Doenças , Quimioterapia Combinada , Feminino , Humanos , Interferon beta/administração & dosagem , Interferon beta/farmacocinética , Neoplasias Renais/tratamento farmacológico , Neoplasias Renais/patologia , Masculino , Melanoma/tratamento farmacológico , Melanoma/patologia , Camundongos , Camundongos Knockout , Mutação , Neoplasias/genética , Polietilenoglicóis/administração & dosagem , Polietilenoglicóis/farmacocinética , Inibidores de Proteínas Quinases/farmacologia , Resultado do Tratamento , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
10.
J Immunol ; 176(4): 2307-15, 2006 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-16455987

RESUMO

The human high m.w.-melanoma-associated Ag (HMW-MAA) is an attractive target for the immunotherapy of melanoma, due to its relatively high expression in a high percentage of melanoma lesions and its restricted distribution in normal tissues. Active immunization with HMW-MAA mimics has been previously shown to induce a HMW-MAA-specific, T cell-dependent Ab response associated with an apparent clinically beneficial effect in advanced melanoma patients. Although T cells play an important role in controlling tumor growth, only limited information is available to date about the induction of HMW-MAA-specific CTL. In this report, we show that immunization of HLA-A2/K(b) transgenic mice with HMW-MAA cDNA-transfected syngeneic dendritic cells elicited a CD8(+) CTL response specific for HMW-MAA peptides with HLA-A2 Ag-binding motifs. The elicited CTL lysed HLA-A2(+)HMW-MAA(+) melanoma cells in vitro, and mouse HLA-A2/K(b) cells pulsed with HMW-MAA-derived peptides in vitro and in vivo. Although this CTL response could be generated in the absence of CD4(+) T cell help, harnessing CD4(+) T cell help in a noncognate Ag-specific manner with the polyclonal activator staphylococcal enterotoxin A augmented the CTL response. These results imply that dendritic cell-based immunization, in combination with CD4(+) T cell help, represents an effective strategy to implement T cell-based immunotherapy targeting HMW-MAA in patients with HMW-MAA-bearing tumors.


Assuntos
Antígenos de Neoplasias/imunologia , Antígenos CD4/imunologia , Linfócitos T CD4-Positivos/imunologia , Antígeno HLA-A2/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Antígenos CD/metabolismo , Antígenos de Diferenciação de Linfócitos T/metabolismo , Linhagem Celular , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Enterotoxinas/farmacologia , Humanos , Imunização , Interferon gama/biossíntese , Lectinas Tipo C , Ativação Linfocitária/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Linfócitos T Citotóxicos/efeitos dos fármacos , Regulação para Cima
11.
Clin Immunol ; 108(1): 8-20, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12865066

RESUMO

Activated T cells with down-regulated L-selectin expression (L-sel(-)) from tumor-draining lymph nodes represent a potent source of specific immune effectors in adoptive immunotherapy. Using congenic pairs of mice and carboxyfluorescein diacetate succinimidyl ester-labeled L-sel(-) T cells, the current study analyzed in vivo proliferation of transferred cells. In the lung of MCA205 tumor-bearing mice, 6% or 0.3 x 10(6) of the 5 x 10(6) donor cells were identified 24 h after transfer. Vigorous proliferation of donor cells was evident on day 2, reaching a maximum on day 6. The proliferation was tumor-specific and CD4 T cells divided with greater magnitude than CD8 cells. Successful adoptive immunotherapy also required sublethal whole-body irradiation (WBI) of the recipient. WBI exerted its effects on facilitating specific T cell proliferation at the tumor site. Taken together, our results demonstrate that adoptively transferred T cells undergo extensive proliferation in response to the tumor and this response is associated with therapeutic efficacy.


Assuntos
Divisão Celular/imunologia , Imunoterapia Adotiva , Linfócitos T/imunologia , Animais , Apresentação de Antígeno , Divisão Celular/efeitos da radiação , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Valor Preditivo dos Testes , Linfócitos T/efeitos da radiação , Irradiação Corporal Total
12.
J Immunol ; 169(9): 4811-21, 2002 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-12391191

RESUMO

Tumor-specific effector T cells (T(E)) are naturally sensitized within the L-selectin(low) (CD62L(low)) fraction of tumor-draining lymph nodes (TDLN). Whether isolated from day 9 (D9) or day 12 (D12) TDLN, 5 million L-selectin(low) T(E) could be culture activated and adoptively transferred to achieve complete rejection of established intradermal, pulmonary, and brain tumors. Surprisingly, although 25 million unfractionated T cells from D9 TDLN were equally effective, even 100 million unfractionated T cells from D12 TDLN seldom prevented lethal intradermal tumor progression, despite a pronounced therapeutic excess of T(E). This highly reproducible treatment failure was due to cotransfer of tumor-induced, L-selectin(high) suppressor T cells (T(S)) which were also present in D12 TDLN. In contrast, D9 TDLN and normal spleens lacked L-selectin(high) T(S). Only those L-selectin(high) D12 TDLN T cells that down-regulated L-selectin during culture activation were suppressive in vivo and in vitro, and, like L-selectin(low) T(E), trafficked promptly into tumors following i.v. administration. This is the first demonstration that adoptive immunotherapy can fail as a direct result of passenger T(S) that share certain phenotypic and trafficking features of T(E), even when otherwise curative doses of T(E) have been administered. Furthermore, in contrast to recently described CD4(+)CD25(+) T(S) and plasmacytoid dendritic cell-activated T(S), tumor-induced L-selectin(high) T(S) prevent tumor rejection via blockade of sensitized, activated T(E) rather than via afferent blockade.


Assuntos
Antígenos de Neoplasias/imunologia , Fibrossarcoma/imunologia , Fibrossarcoma/terapia , Imunoterapia Adotiva/métodos , Selectina L/biossíntese , Linfócitos T Reguladores/imunologia , Animais , Movimento Celular/imunologia , Separação Celular , Progressão da Doença , Regulação para Baixo/imunologia , Feminino , Fibrossarcoma/patologia , Antígenos de Histocompatibilidade/imunologia , Injeções Intradérmicas , Interferon gama/antagonistas & inibidores , Interferon gama/biossíntese , Selectina L/fisiologia , Linfonodos/imunologia , Linfonodos/patologia , Linfonodos/transplante , Ativação Linfocitária , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/patologia , Linfócitos do Interstício Tumoral/transplante , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/patologia , Neoplasias Cutâneas/terapia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/transplante , Linfócitos T Reguladores/metabolismo , Linfócitos T Reguladores/transplante , Falha de Tratamento , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA