Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Xinhai Wan is active.

Publication


Featured researches published by Xinhai Wan.


Clinical Cancer Research | 2012

Modeling a lethal prostate cancer variant with small-cell carcinoma features

Vassiliki Tzelepi; Jiexin Zhang; Jing Fang Lu; Brittany Kleb; Guanglin Wu; Xinhai Wan; Anh Hoang; Kanishka Sircar; Nora M. Navone; Patricia Troncoso; Shoudan Liang; Christopher J. Logothetis; Sankar N. Maity; Ana Aparicio

Purpose: Small-cell prostate carcinoma (SCPC) morphology predicts for a distinct clinical behavior, resistance to androgen ablation, and frequent but short responses to chemotherapy. We sought to develop model systems that reflect human SCPC and can improve our understanding of its biology. Experimental Design: We developed a set of castration-resistant prostate carcinomas xenografts and examined their fidelity to their human tumors of origin. We compared the expression and genomic profiles of SCPC and large-cell neuroendocrine carcinoma (LCNEC) xenografts to those of typical prostate adenocarcinoma xenografts. Results were validated immunohistochemically in a panel of 60 human tumors. Results: The reported SCPC and LCNEC xenografts retain high fidelity to their human tumors of origin and are characterized by a marked upregulation of UBE2C and other mitotic genes in the absence of androgen receptor (AR), retinoblastoma (RB1), and cyclin D1 (CCND1) expression. We confirmed these findings in a panel of samples of CRPC patients. In addition, array comparative genomic hybridization of the xenografts showed that the SCPC/LCNEC tumors display more copy number variations than the adenocarcinoma counterparts. Amplification of the UBE2C locus and microdeletions of RB1 were present in a subset, but none displayed AR nor CCND1 deletions. The AR, RB1, and CCND1 promoters showed no CpG methylation in the SCPC xenografts. Conclusion: Modeling human prostate carcinoma with xenografts allows in-depth and detailed studies of its underlying biology. The detailed clinical annotation of the donor tumors enables associations of anticipated relevance to be made. Future studies in the xenografts will address the functional significance of the findings. Clin Cancer Res; 18(3); 666–77. ©2011 AACR.


Science Translational Medicine | 2014

Prostate cancer cell-stromal cell crosstalk via FGFR1 mediates antitumor activity of dovitinib in bone metastases.

Xinhai Wan; Paul G. Corn; Jun Yang; Nallasivam Palanisamy; Michael W. Starbuck; Elsa M. Li Ning Tapia; Amado J. Zurita; Ana Aparicio; Murali Ravoori; Elba Vazquez; Dan R. Robinson; Yi-Mi Wu; Xuhong Cao; Matthew K. Iyer; Wallace McKeehan; Vikas Kundra; Fen Wang; Patricia Troncoso; Arul M. Chinnaiyan; Christopher J. Logothetis; Nora M. Navone

Dovitinib is therapeutically active in a subset of patients with prostate cancer bone metastases, partly due to blockade of FGFR-mediated stromal-epithelial interactions in the bone microenvironment. Effective to the Bone Bone is the most common site of metastatic spread for prostate cancer, and tumors that have spread to the bone are usually very difficult to treat. Dovitinib is a recently developed drug that inhibits the fibroblast growth factor receptor. Now, Wan et al. have demonstrated that dovitinib is effective for some patients with prostate cancer that has spread to bone. The authors also identified an explanation for some of the observed antitumor effects by showing that the drug interferes with interactions between the prostate cancer cells and surrounding stromal cells in the bone microenvironment. Bone is the most common site of prostate cancer (PCa) progression to a therapy-resistant, lethal phenotype. We found that blockade of fibroblast growth factor receptors (FGFRs) with the receptor tyrosine kinase inhibitor dovitinib has clinical activity in a subset of men with castration-resistant PCa and bone metastases. Our integrated analyses suggest that FGF signaling mediates a positive feedback loop between PCa cells and bone cells and that blockade of FGFR1 in osteoblasts partially mediates the antitumor activity of dovitinib by improving bone quality and by blocking PCa cell–bone cell interaction. These findings account for clinical observations such as reductions in lesion size and intensity on bone scans, lymph node size, and tumor-specific symptoms without proportional declines in serum prostate-specific antigen concentration. Our findings suggest that targeting FGFR has therapeutic activity in advanced PCa and provide direction for the development of therapies with FGFR inhibitors.


Clinical Cancer Research | 2012

Activation of β-Catenin Signaling in Androgen Receptor–Negative Prostate Cancer Cells

Xinhai Wan; Jie Liu; Jing Fang Lu; Vassiliki Tzelepi; Jun Yang; Michael W. Starbuck; Lixia Diao; Jing Wang; Elba Vazquez; Patricia Troncoso; Sankar N. Maity; Nora M. Navone

Purpose: To study Wnt/β-catenin in castrate-resistant prostate cancer (CRPC) and understand its function independently of the β-catenin–androgen receptor (AR) interaction. Experimental Design: We carried out β-catenin immunocytochemical analysis, evaluated TOP-flash reporter activity (a reporter of β-catenin–mediated transcription), and sequenced the β-catenin gene in MDA prostate cancer 118a, MDA prostate cancer 118b, MDA prostate cancer 2b, and PC-3 prostate cancer cells. We knocked down β-catenin in AR-negative MDA prostate cancer 118b cells and carried out comparative gene-array analysis. We also immunohistochemically analyzed β-catenin and AR in 27 bone metastases of human CRPCs. Results: β-Catenin nuclear accumulation and TOP-flash reporter activity were high in MDA prostate cancer 118b but not in MDA prostate cancer 2b or PC-3 cells. MDA prostate cancer 118a and MDA prostate cancer 118b cells carry a mutated β-catenin at codon 32 (D32G). Ten genes were expressed differently (false discovery rate, 0.05) in MDA prostate cancer 118b cells with downregulated β-catenin. One such gene, hyaluronan synthase 2 (HAS2), synthesizes hyaluronan, a core component of the extracellular matrix. We confirmed HAS2 upregulation in PC-3 cells transfected with D32G-mutant β-catenin. Finally, we found nuclear localization of β-catenin in 10 of 27 human tissue specimens; this localization was inversely associated with AR expression (P = 0.056, Fishers exact test), suggesting that reduced AR expression enables Wnt/β-catenin signaling. Conclusion: We identified a previously unknown downstream target of β-catenin, HAS2, in prostate cancer, and found that high β-catenin nuclear localization and low or no AR expression may define a subpopulation of men with bone metastatic prostate cancer. These findings may guide physicians in managing these patients. Clin Cancer Res; 18(3); 726–36. ©2011 AACR.


Bone | 2012

Effect of transforming growth factor beta (TGF-β) receptor I kinase inhibitor on prostate cancer bone growth

Xinhai Wan; Zhi Gang Li; Jonathan M. Yingling; Jun Yang; Michael W. Starbuck; Murali Ravoori; Vikas Kundra; Elba Vazquez; Nora M. Navone

Transforming growth factor beta 1 (TGF-β1) has been implicated in the pathogenesis of prostate cancer (PCa) bone metastasis. In this study, we tested the antitumor efficacy of a selective TGF-β receptor I kinase inhibitor, LY2109761, in preclinical models. The effect of LY2109761 on the growth of MDA PCa 2b and PC-3 human PCa cells and primary mouse osteoblasts (PMOs) was assessed in vitro by measuring radiolabeled thymidine incorporation into DNA. In vivo, the right femurs of male SCID mice were injected with PCa cells. We monitored the tumor burden in control- and LY2109761-treated mice with MRI analysis and the PCa-induced bone response with X-ray and micro-CT analyses. Histologic changes in bone were studied by performing bone histomorphometric evaluations. PCa cells and PMOs expressed TGF-β receptor I. TGF-β1 induced pathway activation (as assessed by induced expression of p-Smad2) and inhibited cell growth in PC-3 cells and PMOs but not in MDA PCa 2b cells. LY2109761 had no effect on PCa cells but induced PMO proliferation in vitro. As expected, LY2109761 reversed the TGF-β1-induced pathway activation and growth inhibition in PC-3 cells and PMOs. In vivo, LY2109761 treatment for 6weeks resulted in increased volume in normal bone and increased osteoblast and osteoclast parameters. In addition, LY2109761 treatment significantly inhibited the growth of MDA PCa 2b and PC-3 in the bone of SCID mice (p<0.05); moreover, it resulted in significantly less bone loss and change in osteoclast-associated parameters in the PC-3 tumor-bearing bones than in the untreated mice. In summary, we report for the first time that targeting TGF-β receptors with LY2109761 can control PCa bone growth while increasing the mass of normal bone. This increased bone mass in nontumorous bone may be a desirable side effect of LY2109761 treatment for men with osteopenia or osteoporosis secondary to androgen-ablation therapy, reinforcing the benefit of effectively controlling PCa growth in bone. Thus, targeting TGF-β receptor I is a valuable intervention in men with advanced PCa.


Oncotarget | 2015

Mature adipocytes in bone marrow protect myeloma cells against chemotherapy through autophagy activation

Zhiqiang Liu; Jingda Xu; Jin He; Huan Liu; Pei Lin; Xinhai Wan; Nora M. Navone; Qiang Tong; Larry W. Kwak; Robert Z. Orlowski; Jing Yang

A major problem in patients with multiple myeloma is chemotherapy resistance, which develops in myeloma cells upon interaction with bone marrow stromal cells. However, few studies have determined the role of bone marrow adipocytes, a major component of stromal cells in the bone marrow, in myeloma chemotherapy resistance. We reveal that mature human adipocytes activate autophagy and upregulate the expression of autophagic proteins, thereby suppressing chemotherapy-induced caspase cleavage and apoptosis in myeloma cells. We found that adipocytes secreted known and novel adipokines, such as leptin and adipsin. The addition of these adipokines enhanced the expression of autophagic proteins and reduced apoptosis in myeloma cells. In vivo studies further demonstrated the importance of bone marrow-derived adipocytes in the reduced response of myeloma cells to chemotherapy. Our findings suggest that adipocytes, adipocyte-secreted adipokines, and adipocyte-activated autophagy are novel targets for combatting chemotherapy resistance and enhancing treatment efficacy in myeloma patients.


Clinical Cancer Research | 2011

Inhibition of prostate cancer osteoblastic progression with VEGF121/rGel, a single agent targeting osteoblasts, osteoclasts, and tumor neovasculature

Khalid A. Mohamedali; Zhi Gang Li; Michael W. Starbuck; Xinhai Wan; Jun Yang; Se Hoon Kim; Wendy Zhang; Michael G. Rosenblum; Nora M. Navone

Purpose: A hallmark of prostate cancer (PCa) progression is the development of osteoblastic bone metastases, which respond poorly to available therapies. We previously reported that VEGF121/rGel targets osteoclast precursors and tumor neovasculature. Here we tested the hypothesis that targeting nontumor cells expressing these receptors can inhibit tumor progression in a clinically relevant model of osteoblastic PCa. Experimental Design: Cells from MDA PCa 118b, a PCa xenograft obtained from a bone metastasis in a patient with castrate-resistant PCa, were injected into the femurs of mice. Osteoblastic progression was monitored following systemic administration of VEGF121/rGel. Results: VEGF121/rGel was cytotoxic in vitro to osteoblast precursor cells. This cytotoxicity was specific as VEGF121/rGel internalization into osteoblasts was VEGF121 receptor driven. Furthermore, VEGF121/rGel significantly inhibited PCa-induced bone formation in a mouse calvaria culture assay. In vivo, VEGF121/rGel significantly inhibited the osteoblastic progression of PCa cells in the femurs of nude mice. Microcomputed tomographic analysis revealed that VEGF121/rGel restored the bone volume fraction of tumor-bearing femurs to values similar to those of the contralateral (non–tumor-bearing) femurs. VEGF121/rGel significantly reduced the number of tumor-associated osteoclasts but did not change the numbers of peritumoral osteoblasts. Importantly, VEGF121/rGel-treated mice had significantly less tumor burden than control mice. Our results thus indicate that VEGF121/rGel inhibits osteoblastic tumor progression by targeting angiogenesis, osteoclastogenesis, and bone formation. Conclusions: Targeting VEGF receptor (VEGFR)-1- or VEGFR-2–expressing cells is effective in controlling the osteoblastic progression of PCa in bone. These findings provide the basis for an effective multitargeted approach for metastatic PCa. Clin Cancer Res; 17(8); 2328–38. ©2011 AACR.


Oncogene | 2016

Hyperactivated FRS2α-mediated signaling in prostate cancer cells promotes tumor angiogenesis and predicts poor clinical outcome of patients.

Junchen Liu; Pan You; Guo Chen; Xin Fu; Xiangfeng Zeng; Cong Wang; Yanqing Huang; Lei An; Xinhai Wan; Nora M. Navone; Chin-Lee Wu; Wallace L. McKeehan; Zhongying Zhang; Weide Zhong; Fen Wang

Metastasis of tumors requires angiogenesis, which is comprised of multiple biological processes that are regulated by angiogenic factors. The fibroblast growth factor (FGF) is a potent angiogenic factor and aberrant FGF signaling is a common property of tumors. Yet, how the aberration in cancer cells contributes to angiogenesis in the tumor is not well understood. Most studies of its angiogenic signaling mechanisms have been in endothelial cells. FGF receptor substrate 2α (FRS2α) is an FGF receptor-associated protein required for activation of downstream signaling molecules that include those in the mitogen-activated protein and AKT kinase pathways. Herein, we demonstrated that overactivation and hyperactivity of FRS2α, as well as overexpression of cJUN and HIF1α, were positively correlated with vessel density and progression of human prostate cancer (PCa) toward malignancy. We also demonstrate that FGF upregulated the production of vascular endothelial growth factor A mainly by increasing expression of cJUN and HIF1α. This then promoted recruitment of endothelial cells and vessel formation for the tumor. Tumor angiogenesis in mouse PCa tissues was compromised by tissue-specific ablation of Frs2α in prostate epithelial cells. Depletion of Frs2α expression in human PCa cells and in a preclinical xenograft model, MDA PCa 118b, also significantly suppressed tumor angiogenesis accompanied with decreased tumor growth in the bone. The results underscore the angiogenic role of FRS2α-mediated signaling in tumor epithelial cells in angiogenesis. They provide a rationale for treating PCa with inhibitors of FGF signaling. They also demonstrate the potential of overexpressed FRS2α as a biomarker for PCa diagnosis, prognosis and response to therapies.


PLOS ONE | 2013

Heme oxygenase-1 (HO-1) expression in prostate cancer cells modulates the oxidative response in bone cells

Mercedes Ferrando; Xinhai Wan; Roberto Meiss; Jun Yang; Adriana De Siervi; Nora M. Navone; Elba Vazquez

Prostate cancer (PCa) is a leading cause of death among males. It is currently estimated that inflammatory responses are linked to 15-20% of all deaths from cancer worldwide. PCa is dominated by complications arising from metastasis to the bone where the tumor cells interact with the bone microenvironment impairing the balance between bone formation and degradation. However, the molecular nature of this interaction is not completely understood. Heme oxygenase-1 (HO-1) counteracts oxidative damage and inflammation. Previous studies from our laboratory showed that HO-1 is implicated in PCa, demonstrating that endogenous HO-1 inhibits bone derived-prostate cancer cells proliferation, invasion and migration and decreases tumor growth and angiogenesis in vivo. The aim of this work was to analyze the impact of HO-1 modulated PCa cells on osteoblasts proliferation in vitro and on bone remodeling in vivo. Using a co-culture system of PC3 cells with primary mice osteoblasts (PMOs), we demonstrated that HO-1 pharmacological induction (hemin treatment) abrogated the diminution of PMOs proliferation induced by PCa cells and decreased the expression of osteoclast-modulating factors in osteoblasts. No changes were detected in the expression of genes involved in osteoblasts differentiation. However, co-culture of hemin pre-treated PC3 cells (PC3 Hem) with PMOs provoked an oxidative status and activated FoxO signaling in osteoblasts. The percentage of active osteoblasts positive for HO-1 increased in calvarias explants co-cultured with PC3 Hem cells. Nuclear HO-1 expression was detected in tumors generated by in vivo bone injection of HO-1 stable transfected PC3 (PC3HO-1) cells in the femur of SCID mice. These results suggest that HO-1 has the potential to modify the bone microenvironment impacting on PCa bone metastasis.


Clinical Cancer Research | 2016

Abstract A03: Analyses of a prostate cancer patient-derived xenografts series, a resource for translational research

Nallasivam Palanisamy; Jun Yang; Xinhai Wan; Elsa M. Li Ning Tapia; John C. Araujo; Estefania Labanca; Louis L. Pisters; Ana Aparicio; Ritu Bhalla; Scott A. Tomlins; Lakshmi P. Kunju; Arul M. Chinnaiyan; Christopher J. Logothetis; Patricia Troncoso; Nora M. Navone

Patients with metastatic prostate cancer (PCa) have effective therapy options, but none of them are curative. Thus, their mortality rates are persistently high. Essential to furthering our progress in PCa research and therapy development is a spectrum of models that reflect the heterogeneity of the disease at each tumor site as well as the different histological variants of PCa (e.g., adenocarcinoma, small cell carcinoma). To address this challenge, we developed a strategy to establish PCa patient-derived xenografts (PDXs), using PCa tissue specimens taken from PCa sites demonstrating clinical progression. This approach provided a diverse repository of PDXs that can be linked prospectively with clinical progression and led to the identification of clinically relevant therapy targets and have proven valuable for testing drugs. We studied the first 50 PDXs developed under our program to a) define the histopathological features of paired human PCa and corresponding PDXs applying the clinically defined morphological characterization groupings of human cancer to the PDX tumors; b) assess the expression of genes known to play roles in PCa pathogenesis (e.g., androgen receptor, PTEN, ETS gene fusions) in PDXs and the human tumors of origin using immunohistochemistry and fluorescence in situ hybridization and c) perform array comparative genomic hybridization to 42 PDXs. We found that the histopathological and molecular pattern of these PDXs maintain the fidelity with the human tumor of origin. Furthermore, of the 50 cases studied, 32 (64%) were adenocarcinomas, and 16 (32%) were small cell carcinomas, poorly differentiated neuroendocrine carcinomas or mixed adenocarcinoma/ small cell carcinomas. In our cohort, we also have one sarcomatoid tumor and one ductal adenocarcinoma. Of the 32 adenocarcinomas in this cohort, 26 were AR-positive (81%), and 11 of the 27 AR-positive adenocarcinomas (41%) had aberrant expression of genes frequently involved in recurrent rearrangement (e.g., ERG, ETV1, ETV5). Also, SCCs and poorly differentiated neuroendocrine carcinomas did not express AR and were negative for ERG. This distribution recapitulates that of human PCa in the general population. Comparative genomic hybridization demonstrated gains and losses previously reported in PCa with a defined cluster of genomic aberrations. Significant differences in oncogenic pathways activation in pairs of PDXs derived from different areas of the same tumor suggesting divergent cellular progression. Finally, using this platform, we identified a focal deletion of speckle-type POZ protein-like (SPOPL) gene in 7/28 PDX. SPOPL is a MATH-BTB protein that shares an overall 85% sequence identity with SPOP (a SPOPL paralog). SPOP was recently reported to be mutated in about 8% of PCa and to define a molecular subclass of PCa. No mutations were found in SPOP in our cohort. In support of our findings, deletions on SPOPL were also found in about 7% of the PCa in TCGA data suggesting that our cohort is a reliable platform for discovery. In conclusion, we have developed a dynamic repository of clinically annotated samples that can be used as a discovery platform. Furthermore, these clinically annotated samples can be linked prospectively to clinical progression/response to therapy and thus will help define therapeutic targets for subpopulations of men and to identify likely responders to previous and upcoming therapies. Citation Format: Nallasivam Palanisamy, Jun Yang, Xinhai Wan, Elsa M. li Ning Tapia, John C. Araujo, Eleni Efstathiou, Estefania Labanca, Louis Pisters, Ana Aparicio, Ritu Bhalla, Scott Tomlins, Lakshmi P. Kunju, Arul Chinnaiyan, Christopher J. Logothetis, Patricia Troncoso, Nora M. Navone. Analyses of a prostate cancer patient-derived xenografts series, a resource for translational research. [abstract]. In: Proceedings of the AACR Special Conference: Patient-Derived Cancer Models: Present and Future Applications from Basic Science to the Clinic; Feb 11-14, 2016; New Orleans, LA. Philadelphia (PA): AACR; Clin Cancer Res 2016;22(16_Suppl):Abstract nr A03.


Cancer Research | 2016

Abstract 1871: Alpha and beta isoforms of fibroblast growth factor receptor 1 in prostate cancer

Estefania Labanca; Xinhai Wan; Jun Yang; Matthew K. Iyer; Christopher J. Logothetis; Arul M. Chinnaiyan; Nora M. Navone

Castrate-resistant progression and bone metastases are hallmarks of advanced prostate cancer (PCa). The fibroblast growth factor (FGF)/FGF receptor (FGFR) complex mediates tumor-stromal interactions and is commonly altered in PCa. FGFR1, FGFR2, FGFR3, and FGFR4 genes encode alternatively spliced variants of FGFRs that vary in the extracellular ligand-binding and intracellular kinase domains. A published study from our group implicated FGFR1 as a therapy target for PCa bone metastases (STM 2014; 6:252ra122). Further, our studies of FGFR1 transcripts by RNA sequencing of 183 human PCas identified eight different protein coding transcripts as the most abundantly expressed with different human PCas expressing different FGFR1 isoforms (Abstract #3913 AACR 2015). These results suggest that different FGFR1 isoforms in PCa cells may partially underlie the biological heterogeneity of PCa. The studies presented here focus in two of the best-characterized isoforms: FGFR1alpha (R1alpha), with 3 Ig-like domains, and 822 aa in length; and FGFR1beta (R1beta), with only 2 Ig-like domains and 733 aa in length. We assessed whether these isoforms induce activation of the same pathways using PC3 cells transiently transfected with empty vector (EV), R1alpha (NM_023110.2) or R1beta (NM_023105.2) and treated with vehicle, FGF2 or FGF9. By Western blot analysis we found that total FGFR1 expression (relative to a loading control) was similar in cells transfected with R1beta or R1alpha. Levels of p-FGFR1 were high in untreated cells transfected with R1alpha, but no further induction was observed after treatment with FGF2 or FGF9. However, p-FGFR1 expression was almost undetectable in untreated cells expressing R1beta and was slightly induced by FGF2 but not by FGF9. p-PLCγ expression was found only in cells expressing R1alpha. We subsequently stably transfected these isoforms in PC3 cells and discovered that while no significant difference in R1alpha and R1beta transcript levels was detected, the levels of R1alpha protein were higher than those of R1beta suggesting that these isoforms may undergo different translational regulation, We also performed in vivo studies by subcutaneous injection of R1alpha or R1beta expressing PC3 cells in immunocompromised mice and weekly monitored tumor volume. We found that PC3 cells expressing R1alpha developed significantly larger tumors than PC3 cells expressing R1beta. We are now in the process of analyzing at the molecular level these tissue specimens. In conclusion, R1alpha and R1beta isoforms trigger different biological effects in PCa cells. Because different isoforms are expressed in different prostate tumors, the expression of these isoforms could be associated with the typical PCa heterogeneity and could explain differences in therapy responses to FGFR1 blockade. These results warrant further studies to fully understand the biological implications of FGFR1 isoforms in the pathogenesis of PCa. Citation Format: Estefania Labanca, Xinhai Wan, Jun Yang, Matthew Iyer, Christopher Logothetis, Arul Chinnaiyan, Nora Navone. Alpha and beta isoforms of fibroblast growth factor receptor 1 in prostate cancer. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 1871.

Collaboration


Dive into the Xinhai Wan's collaboration.

Top Co-Authors

Avatar

Nora M. Navone

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jun Yang

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Christopher J. Logothetis

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Michael W. Starbuck

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Patricia Troncoso

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Elba Vazquez

University of Buenos Aires

View shared research outputs
Top Co-Authors

Avatar

Ana Aparicio

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Estefania Labanca

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge