GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: International Journal of Oncology, Spandidos Publications, ( 2018-03-14)
    Type of Medium: Online Resource
    ISSN: 1019-6439 , 1791-2423
    RVK:
    Language: Unknown
    Publisher: Spandidos Publications
    Publication Date: 2018
    detail.hit.zdb_id: 2079608-0
    detail.hit.zdb_id: 1154403-X
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 7 ( 2022-04-01), p. 1298-1312
    Abstract: Over 50% of all patients with cancer are treated with radiotherapy. However, radiotherapy is often insufficient as a monotherapy and requires a nontoxic radiosensitizer. Squalene epoxidase (SQLE) controls cholesterol biosynthesis by converting squalene to 2,3-oxidosqualene. Given that SQLE is frequently overexpressed in human cancer, this study investigated the importance of SQLE in breast cancer and non–small cell lung cancer (NSCLC), two cancers often treated with radiotherapy. SQLE-positive IHC staining was observed in 68% of breast cancer and 56% of NSCLC specimens versus 15% and 25% in normal breast and lung tissue, respectively. Importantly, SQLE expression was an independent predictor of poor prognosis, and pharmacologic inhibition of SQLE enhanced breast and lung cancer cell radiosensitivity. In addition, SQLE inhibition enhanced sensitivity to PARP inhibition. Inhibition of SQLE interrupted homologous recombination by suppressing ataxia-telangiectasia mutated (ATM) activity via the translational upregulation of wild-type p53-induced phosphatase (WIP1), regardless of the p53 status. SQLE inhibition and subsequent squalene accumulation promoted this upregulation by triggering the endoplasmic reticulum (ER) stress response. Collectively, these results identify a novel tumor-specific radiosensitizer by revealing unrecognized cross-talk between squalene metabolites, ER stress, and the DNA damage response. Although SQLE inhibitors have been used as antifungal agents in the clinic, they have not yet been used as antitumor agents. Repurposing existing SQLE-inhibiting drugs may provide new cancer treatments. Significance: Squalene epoxidase inhibitors are novel tumor-specific radiosensitizers that promote ER stress and suppress homologous recombination, providing a new potential therapeutic approach to enhance radiotherapy efficacy.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16 ( 2020-08-15), p. 3305-3318
    Abstract: There is currently a lack of precise predictive biomarkers for patient selection in clinical trials of inhibitors targeting replication stress (RS) response proteins ATR and CHK1. The objective of this study was to identify novel predictive biomarkers for the response to these agents in treating non–small cell lung cancer (NSCLC). A genome-wide loss-of-function screen revealed that tumor suppressor PPP2R2A, a B regulatory subunit of protein phosphatase 2 (PP2A), determines sensitivity to CHK1 inhibition. A synthetic lethal interaction between PPP2R2A deficiency and ATR or CHK1 inhibition was observed in NSCLC in vitro and in vivo and was independent of p53 status. ATR and CHK1 inhibition resulted in significantly increased levels of RS and altered replication dynamics, particularly in PPP2R2A-deficient NSCLC cells. Mechanistically, PPP2R2A negatively regulated translation of oncogene c-Myc protein. c-Myc activity was required for PPP2R2A deficiency-induced alterations of replication initiation/RS and sensitivity to ATR/CHK1 inhibitors. We conclude that PPP2R2A deficiency elevates RS by upregulating c-Myc activity, rendering cells reliant on the ATR/CHK1 axis for survival. Our studies show a novel synthetic lethal interaction and identify PPP2R2A as a potential new predictive biomarker for patient stratification in the clinical use of ATR and CHK1 inhibitors. Significance: This study reveals new approaches to specifically target PPP2R2A-deficient lung cancer cells and provides a novel biomarker that will significantly improve treatment outcome with ATR and CHK1 inhibitors.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2012
    In:  Cancer Research Vol. 72, No. 3 ( 2012-02-01), p. 666-675
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 3 ( 2012-02-01), p. 666-675
    Abstract: XPC protein is a critical DNA damage recognition factor in nucleotide excision repair for which genetic deficiency confers a predisposition to cancer. In this study, we show that XPC has a function that is independent of its canonical function in DNA repair, potentially altering the interpretation of how XPC deficiency leads to heightened cancer susceptibility. XPC enhances apoptosis induced by DNA damage in a p53 nullizygous background, acting downstream of mitochondrial permeabilization and upstream of caspase-9 activation in the DNA damage–induced apoptosis cascade. We found that deficiency in XPC upregulated production of the short isoform of caspase-2 (casp-2S). This upregulation occurred at both protein and mRNA levels through repression of the caspase-2 promoter by XPC protein. Targeted RNAi-mediated downregulation of casp-2S–enhanced UV-induced apoptosis as well as activation of caspase-9 and caspase-6 in XPC-deficient cells, but not in XPC-proficient cells. In addition, XPC overexpression in various p53-deficient cancer cells resistant to cisplatin improved their sensitivity to cisplatin-induced apoptosis. Given that casp-2S functions as an antiapoptotic protein, our findings suggest that XPC enhances DNA damage–induced apoptosis through inhibition of casp-2S transcription. Together, these findings offer a mechanistic foundation to overcome the resistance of highly prevalent p53-deficient tumors to cell death induced by DNA-damaging therapeutic agents, by targeting strategies that inhibit the expression or function of casp-2S. Cancer Res; 72(3); 666–75. ©2011 AACR.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2012
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 8_Supplement ( 2013-04-15), p. 750-750
    Abstract: Elevated expression of the anti-apoptotic factor Bcl-2 is believed to be one of the contributing factors to an increased relapse rate associated with multiple cisplatin-resistant cancers. DNA damage-binding protein 2 (DDB2) has recently been revealed to play an important role in sensitizing human ovarian cancer cells to cisplatin-induced apoptosis through the down-regulation of Bcl-2, but the underlying molecular mechanism remains poorly defined. Here, we report that DDB2 functions as a transcriptional repressor for Bcl-2. DDB2 binds to a specific response element at the 5’-end of bcl-2 P1 promoter, concomitant with marked losses of histone H3 acetylation along the bcl-2 promoter and enhancer regions. Quantitative chromatin immunoprecipitation analysis revealed that HDAC1 was locally enriched in the bcl-2 P1 core promoter following DDB2 over-expression. Lysine 244 in DDB2 is essential for the transcriptional regulation of bcl-2, evidenced by the fact that the K244E mutant failed to inhibit bcl-2 promoter activity. Interestingly, the simultaneous DDB1 knock-down and DDB2 over-expression are required to potentiate bcl-2 promoter activity in the human ovarian cancer cell line CP70, while the knockdown of DDB1 alone failed to produce any significant effect. Furthermore, quantitative analysis of DDB2 and Bcl-2 expression in ovarian cancers provides clinical evidence that DDB2 and Bcl-2 are inversely correlated. Collectively, our data supports a model in which DDB2 and DDB1 function cooperatively in repressing bcl-2 transcription by recruiting HDAC1 to deacetylate histone H3 across bcl-2 regulatory regions, with DDB2 being responsible for binding to a specific cis-acting element in the bcl-2 P1 promoter. These findings have immediate implications for clinical trial designs of DDB2-based therapies for diverse cisplatin-resistant malignancies.(Supported by NIH grant CA151248 to QEW, and CA93413 to AAW) Citation Format: Ran Zhao, Chunhua Han, Eric Eisenhauer, John Kroger, Li Gong, Qianzheng Zhu, Weiqiang Zhao, Jianhua Yu, Selvendiran Karuppaiyah, Xingluo Liu, Altaf Ahmad Wani, Qi-En Wang. Negative regulation of bcl-2 transcription by DNA damage-binding protein complex in human ovarian cancer cells. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 750. doi:10.1158/1538-7445.AM2013-750
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2013
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2013
    In:  Clinical Cancer Research Vol. 19, No. 19_Supplement ( 2013-10-01), p. A38-A38
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 19, No. 19_Supplement ( 2013-10-01), p. A38-A38
    Abstract: Ovarian cancer is an extremely aggressive disease associated with a high percentage of recurrence and chemotherapy resistance. Damaged DNA binding protein 2 (DDB2) is a 48-kDa protein originally identified as a component of the damage-specific DNA-binding heterodimeric complex DDB. Besides the known ability of DDB2 protein to bind ultraviolet (UV)-damaged DNA and serving as the initial damage recognition factor during nucleotide excision repair (NER). DDB2 was also reported to be involved in the promotion of apoptosis to reduce cisplatin resistance of ovarian cancer cells, control of cell migration, invasiveness and breast tumor progression, as well as suppression of metastasis of colon cancer through a transcriptional regulatory mechanism. Here we show that DDB2 overexpression in human ovarian cancer cell line CP70 suppressed its capability to recapitulate tumors in athymic nude mice, as well as the growth of these cells cultured in semisolid media. Flow cytometry analyses indicate that DDB2 overexpression in CP70 cells reduced the abundance of CD117+ cells and ALDH+ cells, which are believed to be markers of cancer stem cells (CSCs) in ovarian cancer. In contrast, downregulation of DDB2 in ovarian cancer cell line A2780 increased the percentage of ALDH+ cells. In addition, we analyzed the expression level of DDB2 in the putative CSCs isolated from various ovarian cancer cell lines, and demonstrated that DDB2 expression is decreased in the CSCs in comparison with their parental bulk tumor cells. Overexpression of DDB2 in the ovarian CSCs reduced the expression of CSC markers, e.g., Sox2 and Nanog. In addition, overexpression of DDB2 in CSCs isolated from ovarian cancer cell line SKOV-3 also decreased the tumorigenicity of these cells in NOD/SCID mice. In summary, our data suggest that DDB2 suppresses tumorigenicity of ovarian cancer cells through reducing the CSCs pool in cancer cells. Thus, increasing the expression of DDB2 may provide a molecular strategy to inhibit the recurrence of ovarian cancer. (Supported by NIH R01 grant CA151248 to QEW) Citation Format: Chunhua Han, Ran Zhao, John Kroger, Xingluo Liu, Qi-En Wang. DDB2 suppresses tumorigenicity of ovarian cancer cells through reducing the subpopulation of cancer stem cells. [abstract] . In: Proceedings of the AACR Special Conference on Advances in Ovarian Cancer Research: From Concept to Clinic; Sep 18-21, 2013; Miami, FL. Philadelphia (PA): AACR; Clin Cancer Res 2013;19(19 Suppl):Abstract nr A38.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2013
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 25, No. 22_Supplement ( 2019-11-15), p. GMM-017-GMM-017
    Abstract: Cancer stem cells (CSCs) are a particular subpopulation of cells that are characterized by self renewal, differentiation and enhanced tumorigenicity. They are responsible for tumor metastasis, relapse and development of drug resistance. Thus, eradication of CSCs is essential for improved patient prognosis. Micro RNAs are a group of small non-coding, endogenous RNAs that are found to regulate cancer stem cell characteristics by binding to mRNA in a sequence specific manner. In ovarian cancers, a wide array of Micro RNAs have been found to show differential expression of which miR328-3p deserves special mention. In this study, a Micro RNA Nanostring profile analysis reveals a significant upregulation of miR-328-3p in ovarian cancer stem cells isolated from both ovarian cancer cell lines and primary ovarian tumors as compared to their corresponding bulk cells. Moreover, it was found that inhibition of miR-328 limited the CSC population in ovarian cancer cells whereas overexpression of miR-328 enriched the CSC population, thus accounting for miR-328 as an onco-miRNA. The upregulation of miR-328 not only increased the percentage of ALDH+ cells in ovarian cancer bulk cells, but also increased the tumorigenicity and sphere formation ability. This was supported by the orthotopic ovarian xenograft assay. Further investigation revealed that reduced phosphorylation of Erk in ovarian cancer stem cells owing to reduced levels of Reactive Oxygen species (ROS) could be a prospective mechanism behind elevated miR328 expression and maintenance of CSC characteristics. Inhibition of phosphorylated Erk expression in ovarian cancer bulk cells by use of commercially available Erk inhibitor, U0126, led to a significant increase in miR328 expression. Simultaneously, upregulation of phosphorylated Erk in ovarian cancer stem cells not only reduced miR328 expression, but also displayed a significant reduction in expression of cancer stem cell markers (Oct4, Sox2, Nanog), sphere formation ability and tumorigenesis. We obtained a similar trend of results on regulating the expression of pErk by use of Reactive Oxygen Species to ovarian cancer cells. These data further helped us confirm our speculation that reduced ROS promotes the maintenance of CSCs characteristics through inactivation of Erk signalling pathway. Besides, we also identified DDB2 as a direct target of miR328. Our previous findings demonstrate that DDB2 is able to limit ovarian CSC population by disrupting their self renewal capacity. Thus, we conclude that elevated miR328 in ovarian CSCs, resulting from inactivated Erk1/2 activity, is responsible for maintenance of stemness by inhibition of DDB2 expression. Targeting miR-328 could therefore be a novel therapeutic strategy to eradicate CSCs in ovarian cancer. Citation Format: Amit Kumar Srivastava, Tiantian Cui, Ananya Banerjee, Chunhua Han, Shurui Cai, Lu Liu, Dayong Wu1, Ri Cui, Zaibo Li, Xiaoli Zhang, Guozhen Xie, Selvendiran Karuppalyah, Adam Karpf, Jinsong Liu, David Cohn, Qi-En Wang. UPREGULATION OF MIR-328 CONTRIBUTES TO OVARIAN CANCER STEM CELL MAINTENANCE BY DOWNREGULATING DDB2 [abstract]. In: Proceedings of the 12th Biennial Ovarian Cancer Research Symposium; Sep 13-15, 2018; Seattle, WA. Philadelphia (PA): AACR; Clin Cancer Res 2019;25(22 Suppl):Abstract nr GMM-017.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2018
    In:  Clinical Cancer Research Vol. 24, No. 15_Supplement ( 2018-08-01), p. B62-B62
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 24, No. 15_Supplement ( 2018-08-01), p. B62-B62
    Abstract: Cancer stem cells (CSCs), representing the root of many solid tumors including ovarian cancer, have been implicated in disease recurrence, metastasis, and therapeutic resistance. Our previous study has demonstrated that DNA damage-binding protein 2 (DDB2) is able to reduce the abundance of CSCs in the bulk ovarian cancer cells, providing a novel mechanism to explain the DDB2-mediated suppression of tumorigenicity and metastasis, and also suggesting that low expression of DDB2 is required for the maintenance of CSCs. However, the underlying mechanisms still remain unclear. By using the Tet-On DDB2 modulation system, we have confirmed our previous finding that downregulation of DDB2 expands the CSC population in the 2008 ovarian cancer cell line. We also found that DDB2 is able to suppress non-CSC-to-CSC conversions in this cancer cell line. DDB2 has been recognized as a transcriptional regulator. Our microarray analysis has identified ALDH1A1 to be targeted and regulated by DDB2. Downregulation of ALDH1A1 expression by DDB2 at both mRNA and protein levels has been validated in various ovarian cancer cell lines. The mechanistic investigation demonstrated that DDB2 can bind to the promoter region of the ALDH1A1 gene, facilitating the enrichment of histone H3K27me3 by recruiting EZH2 to the promoter region, eventually inhibiting the promoter activity of the ALDH1A1 gene. In addition, we also found that DDB2 competes with transcription factor C/EBPβ for binding to the ALDH1A1 promoter, indirectly inhibiting the ALDH1A1 promoter activity. Finally, we knocked down the expression of DDB2 and ALDH1A1 individually or simultaneously in the 2008 ovarian cancer cell line, and analyzed their tumorigenicity. We found that downregulation of ALDH1A1 is able to block DDB2 silencing-induced expansion of the CSC population, indicating that ALDH1A1 plays a critical role in DDB2-mediated suppression of the CSC population in ovarian cancer cells. We further showed that treatment with a selective ALDH1A1 inhibitor blocked DDB2 silencing-induced expansion of CSCs, and halted orthotopic xenograft tumor growth. In summary, our data demonstrated that DDB2, functioning as a transcription repressor, is able to abrogate ovarian CSC properties by downregulating ALDH1A1 expression. This study provides a novel mechanism underlying the regulation of the CSC population, and would facilitate the development of efficient strategies for eliminating CSCs to prevent tumor relapse and metastasis in ovarian cancers. (Supported by NIH R01CA151248, R01CA211175.) Citation Format: Tiantian Cui, Amit Kumar Srivastava, Chunhua Han, Xiaoli Zhang, Altaf A. Wani, Qi-En Wang. DDB2 represses ovarian cancer cell dedifferentiation by suppressing ALDH1A1. [abstract]. In: Proceedings of the AACR Conference: Addressing Critical Questions in Ovarian Cancer Research and Treatment; Oct 1-4, 2017; Pittsburgh, PA. Philadelphia (PA): AACR; Clin Cancer Res 2018;24(15_Suppl):Abstract nr B62.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2014
    In:  Cancer Research Vol. 74, No. 19_Supplement ( 2014-10-01), p. 2391-2391
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 2391-2391
    Abstract: Ultraviolet light (UV)-induced Xeroderma pigmentosum complementation group C (XPC) protein ubiquitination is mediated by an E3 ubiquitin ligase complex containing UV damaged-DNA binding protein. Here, we report that ubiquitin specific protease 7 (USP7) deubiquitinates XPC during NER. We have demonstrated that transiently compromising cellular USP7, by siRNA or specific inhibitor HBX41108, leads to accumulation of ubiquitinated forms of XPC. However, complete USP7 disruption causes an ubiquitin-mediated XPC degradation upon cellular irradiation. We show that USP7 interacts with XPC in vitro and in vivo. Overexpression of wild-type USP7, but not its catalytically inactive or interaction-defective mutants, reduces ubiquitinated forms of XPC. Importantly, USP7 efficiently deubiquitinates XPC-ubiquitin conjugates in deubiquitination assays in vitro. We further showed that valosin-containing protein (VCP)/p97 is required for UV-induced XPC degradation in USP7-deficient cells. VCP/p97 is readily recruited to DNA damage sites and co-localizes with XPC. Inhibition of VCP/p97 causes an accumulation of ubiquitinated XPC on DNA damaged chromatin. Moreover, USP7 disruption severely impairs the repair of cyclobutane pyrimidine dimers (CPD) and, to a lesser extent, affects the repair of 6-4 photoproducts (6-4PP). Taken together, our findings have uncovered an important role of USP7 in regulating NER via deubiquitinating XPC and by preventing its VCP/p97-regulated proteolysis (This work was supported by grants from NIH). Citation Format: Jinshan He, Qianzheng Zhu, Nidhi Sharma, Gulzar Wani, Chunhua Han, Jiang Qian, Kyle Pentz, Qi-en Wang, Altaf A Wani. USP7 deubiquitinates XPC in response to ultraviolet light irradiation. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 2391. doi:10.1158/1538-7445.AM2014-2391
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2014
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 9 ( 2019-05-01), p. 2314-2326
    Abstract: Cancer stem cells (CSC) play a central role in cancer metastasis and development of drug resistance. miRNA are important in regulating CSC properties and are considered potential therapeutic targets. Here we report that miR-328–3p (miR-328) is significantly upregulated in ovarian CSC. High expression of miR-328 maintained CSC properties by directly targeting DNA damage binding protein 2, which has been shown previously to inhibit ovarian CSC. Reduced activity of ERK signaling in ovarian CSC, mainly due to a low level of reactive oxygen species, contributed to the enhanced expression of miR-328 and maintenance of CSC. Inhibition of miR-328 in mouse orthotopic ovarian xenografts impeded tumor growth and prevented tumor metastasis. In summary, our findings provide a novel mechanism underlying maintenance of the CSC population in ovarian cancer and suggest that targeted inhibition of miR-328 could be exploited for the eradication of CSC and aversion of tumor metastasis in ovarian cancer. Significance: These findings present inhibition of miR-328 as a novel strategy for efficient elimination of CSC to prevent tumor metastasis and recurrence in patients with epithelial ovarian cancer.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...