Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: Breast Cancer Research and Treatment, Springer Science and Business Media LLC, Vol. 201, No. 1 ( 2023-08), p. 43-56
    Abstract: Metastatic hormone receptor-positive (HR+) breast cancer initially responds to serial courses of endocrine therapy, but ultimately becomes refractory. Elacestrant, a new generation FDA-approved oral selective estrogen receptor degrader (SERD) and antagonist, has demonstrated efficacy in a subset of women with advanced HR+breast cancer, but there are few patient-derived models to characterize its effect in advanced cancers with diverse treatment histories and acquired mutations. Methods We analyzed clinical outcomes with elacestrant, compared with endocrine therapy, among women who had previously been treated with a fulvestrant-containing regimen from the recent phase 3 EMERALD Study. We further modeled sensitivity to elacestrant, compared with the currently approved SERD, fulvestrant in patient-derived xenograft (PDX) models and cultured circulating tumor cells (CTCs). Results Analysis of the subset of breast cancer patients enrolled in the EMERALD study who had previously received a fulvestrant-containing regimen indicates that they had better progression-free survival with elacestrant than with standard-of-care endocrine therapy, a finding that was independent estrogen receptor (ESR1) gene mutations. We modeled elacestrant responsiveness using patient-derived xenograft (PDX) models and in ex vivo cultured CTCs derived from patients with HR+breast cancer extensively treated with multiple endocrine therapies, including fulvestrant. Both CTCs and PDX models are refractory to fulvestrant but sensitive to elacestrant, independent of mutations in ESR1 and Phosphatidylinositol-4,5-Bisphosphate 3-Kinase Catalytic Subunit Alpha ( PIK3CA) genes. Conclusion Elacestrant retains efficacy in breast cancer cells that have acquired resistance to currently available ER targeting therapies. Elacestrant may be an option for patients with HR+/HER2- breast cancer whose disease progressed on fulvestrant in the metastatic setting. Translational Relevance. Serial endocrine therapy is the mainstay of management for metastatic HR+breast cancer, but acquisition of drug resistance highlights the need for better therapies. Elacestrant is a recently FDA-approved novel oral selective estrogen receptor degrader (SERD), with demonstrated efficacy in the EMERALD phase 3 clinical trial of refractory HR+breast cancer. Subgroup analysis of the EMERALD clinical trial identifies clinical benefit with elacestrant in patients who had received prior fulvestrant independent of the mutational status of the ESR1 gene, supporting its potential utility in treating refractory HR+breast cancer. Here, we use pre-clinical models, including ex vivo cultures of circulating tumor cells and patient-derived xenografts, to demonstrate the efficacy of elacestrant in breast cancer cells with acquired resistance to fulvestrant.
    Type of Medium: Online Resource
    ISSN: 0167-6806 , 1573-7217
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2023
    detail.hit.zdb_id: 2004077-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Letters, Elsevier BV, Vol. 371, No. 2 ( 2016-02), p. 326-333
    Type of Medium: Online Resource
    ISSN: 0304-3835
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2016
    detail.hit.zdb_id: 195674-7
    detail.hit.zdb_id: 2004212-7
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Discovery, American Association for Cancer Research (AACR), Vol. 11, No. 3 ( 2021-03-01), p. 678-695
    Abstract: Circulating tumor cells (CTC) are shed by cancer into the bloodstream, where a viable subset overcomes oxidative stress to initiate metastasis. We show that single CTCs from patients with melanoma coordinately upregulate lipogenesis and iron homeostasis pathways. These are correlated with both intrinsic and acquired resistance to BRAF inhibitors across clonal cultures of BRAF-mutant CTCs. The lipogenesis regulator SREBP2 directly induces transcription of the iron carrier Transferrin (TF), reducing intracellular iron pools, reactive oxygen species, and lipid peroxidation, thereby conferring resistance to inducers of ferroptosis. Knockdown of endogenous TF impairs tumor formation by melanoma CTCs, and their tumorigenic defects are partially rescued by the lipophilic antioxidants ferrostatin-1 and vitamin E. In a prospective melanoma cohort, presence of CTCs with high lipogenic and iron metabolic RNA signatures is correlated with adverse clinical outcome, irrespective of treatment regimen. Thus, SREBP2-driven iron homeostatic pathways contribute to cancer progression, drug resistance, and metastasis. Significance: Through single-cell analysis of primary and cultured melanoma CTCs, we have uncovered intrinsic cancer cell heterogeneity within lipogenic and iron homeostatic pathways that modulates resistance to BRAF inhibitors and to ferroptosis inducers. Activation of these pathways within CTCs is correlated with adverse clinical outcome, pointing to therapeutic opportunities. This article is highlighted in the In This Issue feature, p. 521
    Type of Medium: Online Resource
    ISSN: 2159-8274 , 2159-8290
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    detail.hit.zdb_id: 2607892-2
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Breast Cancer Research and Treatment, Springer Science and Business Media LLC, Vol. 188, No. 1 ( 2021-07), p. 43-52
    Type of Medium: Online Resource
    ISSN: 0167-6806 , 1573-7217
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2021
    detail.hit.zdb_id: 2004077-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Nature Genetics, Springer Science and Business Media LLC, Vol. 49, No. 1 ( 2017-1), p. 65-74
    Type of Medium: Online Resource
    ISSN: 1061-4036 , 1546-1718
    RVK:
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2017
    detail.hit.zdb_id: 1494946-5
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2023
    In:  Cancer Research Vol. 83, No. 7_Supplement ( 2023-04-04), p. 1668-1668
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 1668-1668
    Abstract: A subset of tumor cells escapes apoptotic and necroptotic death when subjected to chemo- or targeted therapy thereby entering a transient reversible less-proliferative “tolerant state.” Upon continuous drug treatment, these tumor cells become drug resistant, leading to metastases and cancer relapse. A long-standing goal in oncology has been to develop therapeutics to effectively target both cell states. Growing published evidence suggests that accumulation of reactive oxygen species and lipid remodeling make tolerant and resistant tumor cells more vulnerable to ferroptotic mode of cell death. To this end, we here characterize the activity of a proprietary inhibitor of GPX4, an enzyme that safeguards against ferroptosis, across a panel of tolerant and resistant cell line models. Drug tolerant and resistant cancer cell lines spanning three indications were generated by applying continuous treatments with relevant standard-of-cares (SOC) in vitro for short or long durations, respectively. Age-matched control cell lines were derived in parallel by treatment of parental cells with DMSO. Cells were subjected to high dose of SOC for nine days to generate tolerant lines, or to increased therapeutic doses of SOC over three months to generate resistant lines, after which resistance to SOCs was confirmed. In accordance with our hypothesis, we found that drug tolerant lines demonstrated enhanced sensitivity to GPX4 inhibition compared to age-matched control lines. Furthermore, we noted that resistant tumor cell lines retained exquisite sensitivity to GPX4 inhibition across all indications while demonstrating up to 1000-fold resistance to SOCs. In summary, we found that cell line models which evade cell death upon treatment with chemo- or targeted therapies continued to present vulnerability to ferroptosis induction across both cell states. Given the heterogeneity in responses to drug-induced cell-states, understanding the mechanisms of differential drug sensitivity to GPX4 inhibition may help in formulating successful strategies for targeting this relevant cell population in the clinic, thereby limiting cancer progression and/or improving curative potential. Citation Format: Taronish Dubash, Maria Cristina Munteanu, Shrouq Farah, Cristian Nunez, Laurence Jadin, Branko Radetich, Darby Schmidt, Nadia Gurvich. Targeting drug-induced tolerant & resistant cell populations by novel ferroptosis inducer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 1668.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 4_Supplement ( 2022-02-15), p. PD2-03-PD2-03
    Abstract: Background: For patients with hormone receptor-positive (HR+) metastatic breast cancer (MBC) and progression on combination endocrine therapy plus palbociclib/ribociclib, abemaciclib can be effective and well-tolerated, conferring durable clinical benefit in a subset of patients (Wander SA et al. JNCCN 2021). We previously reported that, even in patients with ESR1-mutant (ESR1-MUT) HR+ MBC, some - but not all - still achieve long-term disease control with abemaciclib (Wander SA et al. SABCS 2020). How to identify which patients are likely to benefit from abemaciclib after progression on combination endocrine therapy and CDK4/6 inhibition is not known and, more generally, predicting tumor response to CDK4/6 inhibition has been an area of ongoing research. Because resistance to CDK4/6 inhibition can occur through multiple mechanisms, we hypothesized that a comprehensive resistance panel, rather than single genetic markers or immunohistochemical readouts, would provide an effective predictive tool. Methods: To determine which patients with ESR1-MUT MBC and progression on endocrine therapy plus palbociclib/ribociclib benefit from abemaciclib, we examined a multicenter cohort of such patients who had genomic profiling by standard commercially available assays, the majority of which were via plasma-based cell-free DNA (cfDNA) genotyping assays. We generated a curated list based upon prior literature of CDK4/6i resistance drivers that had been validated in tumor biopsy specimens and in laboratory models: these genes were involved in cell cycle regulation (CCNE1/2, RB1, AURKA) and growth factor signaling pathways (ERBB2, FGFR1/2, AKT1, PTEN, KRAS, FAT1). Progression-free survival (PFS) was defined as time from abemaciclib initiation to time of discontinuation due to disease progression or death; patients who discontinued due to toxicity were right-censored. To examine the cellular effects of different mutations, we also studied the impact of ESR1-MUT, RB1 loss, and KRAS activation on the growth and survival (using an ATP abundance-based assay) of patient-derived circulating tumor cell (CTC) lines treated with palbociclib and abemaciclib in vitro. Results: Among patients with ESR1-MUT MBC with disease progression on endocrine therapy plus palbociclib/ribociclib (n=28), absence of co-existing genomic alterations in our curated panel was associated with greater clinical benefit with subsequent abemaciclib. Patients lacking a mutation in this resistance panel (n=17) had a median PFS of 7.0 months (95% CI: 4.1-13.2); patients with at least one mutation in this panel (n=11) had a median PFS of 3.5 months (95% CI: 2.1-5.4). The difference in PFS was statistically significant (p=0.02, log-rank test). On univariable Cox regression the hazard ratio for patients with a mutation in the resistance panel was 2.8 (95% CI: 1.1-7.1, p=0.03). In vitro, two out of three patient-derived cell lines with ESR1-MUT remained sensitive to abemaciclib, while those with mutation in RB1 or KRAS were less sensitive to abemaciclib. Conclusions: In our study, absence of co-existing genomic alterations in a curated panel was associated with greater clinical benefit with subsequent abemaciclib among patients with ESR1-MUT MBC with prior disease progression on endocrine therapy plus palbociclib/ribociclib. While a small dataset, this is the first demonstration of a genomic panel associated with continued CDK4/6 inhibitor sensitivity. Future directions include testing this panel outside of ESR1-MUT MBC and refining the panel in additional datasets with increased sample size, to guide therapy selection for patients with HR+ MBC. Citation Format: Jamie O Brett, Taronish D Dubash, Andrzej Niemierko, Veronica Mariotti, Leslie SL Kim, Jing Xi, Apurva Pandey, Siobhan Dunne, Azadeh Nasrazadani, Maxwell R Lloyd, Laura M Spring, Douglas Micalizzi, Maristela Onozato, Dante Che, Adam Brufsky, Kevin M Kalinsky, Cynthia X Ma, Joyce O’Shaughnessy, Hyo S Han, A. John Iafrate, Shyamala Maheswaran, Daniel A Haber, Aditya Bardia, Seth A Wander. Association between co-existing genomic alterations and abemaciclib benefit in patients with metastatic hormone receptor-positive breast cancer with ESR1 mutations following disease progression on prior endocrine therapy plus palbociclib or ribociclib [abstract]. In: Proceedings of the 2021 San Antonio Breast Cancer Symposium; 2021 Dec 7-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2022;82(4 Suppl):Abstract nr PD2-03.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 3043-3043
    Abstract: Metastasis formation is the most common cause of colorectal cancer (CRC) related death. How cancer cells disseminate from the primary tumor, gain access to blood and lymphatic vessels and seed to distant organs is not fully understood. To gain deeper mechanistic insights into metastasis formation, we developed a metastasis xenotransplantation model for primary human colorectal cancer cells. Patient (n=12) derived tumor-initiating cells (TIC) were enriched in spheroid cultures and subsequently transplanted into the kidney capsule of immunodeficient NOD/SCID-IL2RGnull (NSG) mice. Importantly, cultures derived from 3 distinct patients frequently metastasized into the murine liver and spleen, whereas spheroids derived from all other patients (n=9) never did. Compared to non-metastasizing spheroids, metastasizing spheroids strongly expressed mesenchymal markers (e.g. Vimentin and N-cadherin), whereas epithelial and colonic differentiation markers (e.g. E-cadherin, Trefoil factor 3, Defensin alpha 5, Keratin-8, -18 -20) were down-regulated suggesting that metastasizing primary patient derived CRC spheroids may have passed through an epithelial to mesenchymal transition (EMT). As the miR-200 family has been shown to support an epithelial phenotype and has been implicated in the process of EMT in tumor cells, we analyzed miRNA expression in metastasizing versus non-metastasizing spheroid cultures. Interestingly, metastasizing spheroids strongly down-regulated the miR-200 clusters miR-200a/-200b/-429 and miR-200c/-141 as well as miR-194 and miR-203. To further understand the mechanism of deregulated gene- and miRNA expression in metastasizing spheroids, we evaluated the DNA methylation status of miRNA promoters. In contrast to non-metastasizing spheroid cultures, metastasizing spheroids showed pronounced DNA methylation within the miR-200 promoter regions, indicating that miR-200 expression in primary metastasizing human colorectal cancer spheroids is suppressed through epigenetic mechanisms. Forced overexpression of both miR-200 clusters in metastasizing spheroids restored expression of epithelial genes like E-Cadherin and Keratin-8 and reduced expression of the mesenchymal markers Vimentin and N-cadherin. Moreover, incubation with low dose 5-aza-2′-deoxycytidine increased the expression of epithelial cell adhesion molecule (EpCAM) in metastasizing spheroids, indicating a partial reversion of the mesenchymal phenotype by demethylating agents. In summary, our findings indicate that metastasizing primary human CRC spheroids are epigenetically fixed in a mesenchymal state. This model allows further dissecting and understanding mechanisms of EMT and metastasis formation in human CRC thereby providing the basis for the development of future therapeutic intervention strategies against metastasizing human colorectal cancer. Citation Format: Christopher M. Hoffmann, Klara M. Giessler, Claudia R. Ball, Taronish D. Dubash, Sebastian M. Dieter, Sarah Bergmann, Wilko Weichert, Christof Von Kalle, Martin Schneider, Constance Baer, Christoph Plass, Manfred Schmidt, Hanno Glimm. Mesenchymal phenoptype of metastasizing primary human colorectal TIC is maintained through epigenetic silencing of miR-200. [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 3043. doi:10.1158/1538-7445.AM2014-3043
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 771-771
    Abstract: Detection of multiple primary lung cancers is increasing in frequency, with up to 15% of all non-small cell lung cancer (NSCLC) patients presenting with two or more anatomically separate tumor nodules on CT scans. Increased detection is in part due to expanded lung cancer screening criteria in an aging population. Distinguishing multiple primary tumors from intrapulmonary metastases can be challenging, yet is critical for determining clinical management. Current models predict development of multiple primary tumors out of a cancerized field, such as occurs due to smoking. The occurrence of multiple primary tumors is unexplained in patients with EGFR-mutant lung cancer (~15% of all NSCLC), lacking environmental exposures. We identified patients with non-small cell lung cancer (NSCLC) who presented with multiple primary EGFR-mutant tumors, in the absence of family history of lung cancer or heavy smoking. We subjected the macrodissected tumors and surrounding normal tissues to whole exome sequencing as well as to analysis of hypermutable poly-guanine (poly-G) repeats, which are two orthogonal methods of lineage tracing. An additional familial case with a germline EGFR-T790M mutation was used to establish parameters for timing of somatic mutations, and functional properties of novel germline variants were modeled in vitro. Of eleven non-familial NSCLC cases with two or more geographically distinct EGFR-mutant lung cancers, two patients harbored a germline EGFR variant allele, which confers moderately enhanced signaling in vitro, followed by a somatically acquired EGFR mutation. In an additional four cases, both whole exome sequencing and poly-G repeat analyses indicate a distant shared somatic cell-of-origin, consistent with embryonic mosaicism. Three cases revealed clinically unappreciated metastasis and two cases remain unexplained. Together, our data suggest that multiple primary lung cancers with somatic EGFR driver mutations may result from genetic susceptibility, attributable either to attenuated EGFR germline variants or to embryonic mosaicism resulting in geographically disparate patches of predisposed lung tissue. Such predisposed cases should be surveilled for early detection of future tumors, and surgical resection in these cases should consider the life-long risk of additional cancers. Citation Format: Risa Burr, Ignaty Leshchiner, Christina L. Costantino, Martin Blohmer, Tilak Sundaresan, Justin Cha, Karsen Seeger, Sara Guay, Brian P. Danysh, Ira Gore, Raquel A. Jacobs, Kara Slowik, Filippo Utro, Kahn Rhrissorrakrai, Chaya Levovitz, Jaimie L. Barth, Taronish Dubash, Brian Chirn, Laxmi Parida, Lecia V. Sequist, Mari Mino-Kenudson, Kamila Naxerova, Shyamala Maheswaran, Gad Getz, Daniel A. Haber. Mechanisms of genetic predisposition to multifocal lung cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 771.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 14_Supplement ( 2016-07-15), p. 910-910
    Abstract: A subpopulation of tumor-initiating cells (TIC) drives colorectal cancer (CRC) progression in serial xenotransplantation. Strikingly, the CRC TIC compartment itself is heterogeneous and comprised of a hierarchy of long-term (LT-) TIC, tumor transient amplifying cells (T-TAC) and delayed contributing (DC-) TIC. Whether this functionally heterogeneous TIC compartment is genetically homogenous or whether distinct genetic subclones drive the functional heterogeneity of the TIC compartment is yet unknown. To address this question, we performed high coverage (91-126 fold) whole genome sequencing on primary CRC patient tumors (n = 3), corresponding serially passaged TIC enriched spheroids as well as spheroid derived serial xenografts. Sequenced samples harbored between 22.800 and 232.000 synonymous or non-synonymous single nucleotide variants (SNVs). In addition, all samples contained multiple focal or large-scale somatic copy number alterations (CNAs). Clustering of SNVs as well as subclonal copy numbers from serial xenografts and spheroids were used to define SNV- and CNA-based subpopulations. Next, cellular fractions of identified subpopulations were determined and combined applying maximal parsimony to create models of the minimal number of subclones present in each sample. Using this strategy, we found that multiple subclones were present in each sample analyzed. Subclone heterogeneity was maintained during serial in vitro passaging and serial xenografting. Importantly, tumor initiation in xenografts was driven by at least 3 distinct genetic subclones whose relative contribution dynamically changed over time. Strikingly, in serial samples from the same patients, different genetic subclones grew out in vivo and in vitro. To test whether functional heterogeneity of the TIC compartment is related to the presence of genetic subclones, we assessed the contribution of different TIC subtypes - LT-TIC, T-TAC and DC-TIC - at early and late time points of xenografting using secondary genetic marking. Therefore, 1×10⁁5 cells derived from early and late generation xenografts were transduced with an integrating lentiviral vector, thereby generating a stable barcode-like genetic mark which differs in each transduced cell. Following serial transplantation of transduced cells for 3 mouse generations, tumors were harvested and lentiviral integration sites were determined using highly sensitive LAM-PCR and high-throughput sequencing. Assessing the relative contribution of LT-TIC, T-TAC and DC-TIC revealed that the functional heterogeneity of the TIC compartment was preserved despite profound genetic subclone dynamics in serial xenotransplantation. These results strongly indicate that multiple genetic subclones drive long-term tumor formation and that functional heterogeneity of the CRC TIC compartment is not based on specific genetic subclones. Citation Format: Klara M. Giessler, Kortine Kleinheinz, Gnana Prakash Balasubramanian, Daniel Hübschmann, Taronish D. Dubash Rai, Sebastian D. Dieter, Christine Siegl, Christopher M. Hoffmann, Sarah Weber, Raffaele Fronza, Saira Afzal, Manfred Schmidt, Martin Schneider, Alexis Ulrich, Juergen Weitz, Wilko Weichert, Matthias Schlesner, Benedikt Brors, Claudia R. Ball, Hanno Glimm. Genetic subclone heterogeneity of the human colon cancer initiating cell compartment. [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 910.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages