Skip to main content
Erschienen in: Journal of Hematology & Oncology 1/2015

Open Access 01.12.2015 | Case report

Comprehensive serial molecular profiling of an “N of 1” exceptional non-responder with metastatic prostate cancer progressing to small cell carcinoma on treatment

verfasst von: Kunal C. Kadakia, Scott A. Tomlins, Saagar K. Sanghvi, Andi K. Cani, Kei Omata, Daniel H. Hovelson, Chia-Jen Liu, Kathleen A. Cooney

Erschienen in: Journal of Hematology & Oncology | Ausgabe 1/2015

Abstract

Importance

Small cell carcinoma/neuroendocrine prostate cancer (NePC) is a lethal, poorly understood prostate cancer (PCa) subtype. Controversy exists about the origin of NePC in this setting.

Objective

To molecularly profile archived biopsy specimens from a case of early-onset PCa that rapidly progressed to NePC to identify drivers of the aggressive course and mechanisms of NePC origin and progression.

Design, setting, and participants

A 47-year-old patient presented with metastatic prostatic adenocarcinoma (Gleason score 9). After a 6-month response to androgen deprivation therapy, the patient developed jaundice and liver biopsy revealed exclusively NePC. Targeted next generation sequencing (NGS) from formalin-fixed paraffin-embedded (FFPE)-isolated DNA was performed from the diagnostic prostate biopsy and the liver biopsy at progression.

Intervention

Androgen deprivation therapy for adenocarcinoma followed by multiagent chemotherapy for NePC.

Main outcomes and measures

Identification of the mutational landscape in primary adenocarcinoma and NePC liver metastasis. Whether the NePC arose independently or was derived from the primary adenocarcinoma was considered based on mutational profiles.

Results

A deleterious somatic SMAD4 L535fs variant was present in both prostate and liver specimens; however, a TP53 R282W mutation was exclusively enriched in the liver specimen. Copy number analysis identified concordant, low-level alterations in both specimens, with focal MYCL amplification and homozygous PTEN, RB1, and MAP2K4 losses identified exclusively in the NePC specimen. Integration with published genomic profiles identified MYCL as a recurrently amplified in NePC.

Conclusions and relevance

NGS of routine biopsy samples from an exceptional non-responder identified SMAD4 as a driver of the aggressive course and supports derivation of NePC from primary adenocarcinoma (transdifferentiation).
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​s13045-015-0204-7) contains supplementary material, which is available to authorized users.
Kunal C. Kadakia and Scott A. Tomlins contributed equally to this work.

Competing interests

The University of Michigan has been issued a patent on the detection of ETS gene fusions in prostate cancer, on which S.A.T is a co-inventor. The University of Michigan licensed the diagnostic field of use to Gen-Probe, Inc., who has sublicensed some rights to Ventana/Roche. S.A.T. serves as a consultant to, and has received honoraria from, Ventana/Roche. No other disclosures are reported.

Authors’ contributions

KK and ST contributed equally to the study. ST participated in the conception, design, funding, study supervision, and carried out the molecular genetic studies, participated in the analysis and interpretation of the data as well as drafting, and critical review of the manuscript. KK participated in the conception, design, analysis, and interpretation of the data, as well as drafting and critical review of the manuscript. SS participated in the analysis and interpretation of the data and drafting of the manuscript. AC, KO, DH, CL all participated in the acquisition, analysis, and interpretation of the data as well as the administrative, technical, and material support. DH also performed the statistical analysis. KC participated in the conception, design, funding, and study supervision, and participated in the analysis and interpretation of the data as well as drafting and critical review of the manuscript. All authors read and approved the final manuscript.

Introduction

Precision oncology heralds an era in which tumors are biopsied and profiled in the metastatic setting with the goal of identifying therapeutic targets. Although next generation sequencing (NGS) of “N of 1” cases have identified mechanisms of exceptional response to investigational therapies [14], such approaches have largely not been applied to exceptional non-responders. Likewise, NGS profiling of pre-/post-treatment samples in cases with marked histologic progression, which enables assessment of progression mechanisms, is challenging due to difficulties in obtaining and assessing routine diagnostic biopsy samples. Here, we describe NGS assessment of routine clinical samples from a patient diagnosed with metastatic PCa at a young age who rapidly progressed and died from disease approximately 1 year from diagnosis. Importantly, while his primary tumor exclusively contained conventional prostatic adenocarcinoma, a post-treatment liver metastasis biopsy exclusively contained prostatic small cell carcinoma/NePC. Hence, this case provided a unique opportunity to assess the utility of NGS-based profiling of serial routine biopsy specimens from an “exceptional non-responder” who showed rapid histologic progression during treatment.

Methods

Study oversight

The patient signed a consent form to participate in an IRB-approved research study to sequence tumor and germline DNA from men presenting with metastatic PCa before age 60 years.

Tumor sequencing and analysis

Post-mortem, we performed targeted next generation sequencing (NGS) on DNA and RNA co-isolated from macrodissected formalin-fixed paraffin-embedded (FFPE) tissue sections from the original diagnostic prostate biopsy specimen (PR-259) and the post-treatment liver biopsy specimen (PR-258). Multiplexed PCR-based NGS (Ampliseq) was performed using 40-ng DNA and the Ion Torrent Comprehensive Cancer Panel (CCP), which targets the coding region of 409 cancer-related genes with 15,992 amplicons (1,688,650 targeted bases) [5, 6]. Multiplexed PCR-based NGS (Ampliseq) was also performed using 20 ng RNA and the RNA component of the Oncomine Comprhensive Panel (OCP), which uses a total of 154 primer pairs to target known gene fusion isoforms, including those involving recurrent 5’ (TMPRSS2, SLC45A3, C15ORF21) and 3’ (ERG, ETV1, ETV4, ETV5, and BRAF) fusion partners in PCa [7]. Detailed description of sequencing, data analysis using validated pipelines, sequencing statistics, and all identified high-confidence somatic variants are given in the Supplement (Additional file 1 and Tables 1 and 2).
Table 1
Sequencing statistics for the diagnostic prostate biopsy sample containing conventional adenocarcinoma (PR-259) and subsequent liver metastasis with small cell/neuroendocrine carcinoma (PR-258)
Parameter
PR259
PR258
DNA sequencing
  
 Mapped reads (n)
25,670,652
2,937,737
 On target reads (%)
98.1 %
99.0 %
 Total aligned base reads
2,672,758,224
322,308,287
 Total base reads on target
2,562,288,371
310,618,776
 Average base coverage depth
1,517
184
 Uniformity of base coverage
51.2 %
90.9 %
 Target base coverage at 20×
89.9 %
94.2 %
 Target base coverage at 100×
71.7 %
72.4 %
 Target bases with no strand bias
93.5 %
93.3 %
 Total called variantsa
2,556
1,177
 Variants passing filteringb
5
6
 Somatic variantsc
3
4
 Prioritized somatic variantsd
1
2
RNA sequencing
  
 Total reads (n)
66,564
247,655
 Uniquely mapped to genome (%)
38 %
79 %
 Identified gene fusions (n)
0
0
aVariants called by automated low stringency variant calling
bVariants passing filtering of technical artifacts, poorly supported variants, germline SNPs and synonymous/non-coding variants
cVariants confirmed as somatic through exome sequencing of germline DNA
dSomatic variants prioritized as likely driving oncogenic or tumor suppressive mutations as described in the eMethods
Table 2
High confidence, non-synonymous variants identified in the diagnostic prostate biopsy sample containing conventional adenocarcinoma (Dx [PR-259]) and subsequent liver metastasis with small cell/neuroendocrine carcinoma (NePC [PR-258])
     
Dx (PR-259)
NePC (PR-258)
  
Location
Gene
Ref
Alt
AA change
Var. allele frequency (FAO/FDP)
Read depth (FDP)
Var. allele frequency (FAO/FDP)
Read depth (FDP)
AV_TX
AV_NUC
chr17:7577094
TP53
G
A
p.R282W
1.6 %
755
69 %
217
NM_000546
c.C844T
chr18:48604783
SMAD4
C
p.L535fs
36 %
227
67 %
196
NM_005359
c.1605delC
chr12:49449077
KMT2D
T
C
p.K11E
18 %
22
37 %
180
NM_003482
c.A31G
chr5:7878077
MTRR
C
G
p.P141R
15 %
89
55 %
142
NM_002454
c.C422G
chr19:17953318
JAK3
C
T
p.R223H
54 %
76
60 %
70
NM_000215
c.G668A
chr6:32166327
NOTCH4
T
C
p.T1543A
42 %
608
35 %
231
NM_004557
c.A4627G
High confidence, non-synonymous variants identified in the diagnostic prostate biopsy sample containing conventional adenocarcinoma (Dx [PR-259]) and paired subsequent liver metastasis with small cell/neuroendocrine carcinoma (NePC [PR-258]) are shown. For each variant, the location (hg19), gene, reference (Ref.) and variant (Alt.) alleles, and amino acid (AA) change info is given. The variant (Var.) allele frequency is the flow-corrected variant containing read count (FAO) divided by flow-corrected read depth (FDP). For the KMT2D and TP53 variants in PR-259, the Var allele frequency was calculated using non-flow-corrected read counts (AO/DP) due to those variants not passing filtering in that sample. The reference sequence (Refseq) and nucleotide (Nuc) change used to derive the AA change are also given. Variants identified as SNPs by exome sequencing of germline DNA are indicated in gray. Prioritized somatic variants are bolded

Case presentation

A 47-year-old male participated in PCa screening due to positive family history of PCa (father). Prostate-specific antigen (PSA) was initially elevated at 13.3 ng/mL, however, prostate biopsy was negative. His PSA rose one year later to 170 ng/mL, and repeat prostate biopsy revealed Gleason 4 + 5 = 9 prostate adenocarcinoma involving all 12 cores (Fig. 1, top and middle panels). Computed tomography (CT) of the abdomen and pelvis showed an enlarged left iliac lymph node but no other metastases. Bone-scan showed metastases involving the lumbar vertebrae. He enrolled in a clinical trial and was treated with an oral anti-androgen along with leuprolide. On this regimen, PSA decreased to a nadir of <4.0 ng/mL over 6 months, and CT scan showed reduction in the size of the enlarged iliac node and normal liver parenchyma.
Two months later, the patient presented with jaundice and was found to have elevated transaminases. MRI showed a pseudocirrhotic appearance of the liver (Fig. 2). Liver biopsy revealed small cell/NePC (Fig. 1, bottom panel). Despite poor performance status and after discussion of risks and benefits, he was initiated on dose-reduced oral etoposide along with carboplatin and continued leuprolide. Although the patient demonstrated an initial clinical response, he elected to end treatment. He died under hospice care 3 months following NePC diagnosis.

Results

NGS of DNA isolated from the routine FFPE diagnostic prostate biopsy specimen (PR-259) and the post-treatment liver biopsy specimen (PR-258) identified a total of two prioritized high-confidence somatic variants. As shown in Fig. 3, a SMAD4 c1605delC p.L535fs frameshifting variant was present in both PR-259 (36 % variant allele frequency) and PR-258 (67 % variant allele frequency). In contrast, a TP53 c.C844T p.R282W non-synonymous variant was exclusively called in the NePC specimen (PR-258; 69 % variant allele frequency). This variant was markedly enriched in PR-258, and was only present at a variant allele frequency of 1.6 % (12/755 reads) in the diagnostic pre-treatment specimen (PR-259). These results are consistent with clonal origin and marked enrichment of the TP53 R282W variant exclusively in the post-treatment NePC specimen. Exome sequencing of germ line DNA isolated from white blood cells confirmed the TP53 and SMAD4 variants as somatic (see Table 2).
We next compared NGS-derived copy number profiles between PR-259 and PR-258 using our well-validated approach [57]. Copy number profiling revealed broad one copy loss of 10q (containing PTEN), 18q, and a complex alteration on chromosome 19 in both specimens, whereas the post-treatment liver biopsy (PR-258) exclusively demonstrated focal, high-level MYCL amplification, and focal homozygous PTEN, RB1, and MAP2K4 deletions. Lastly, no gene fusions were identified in either PR-259 or PR-258 from targeted multiplexed PCR-based RNAseq on co-isolated RNA (see Additional file 1). Taken together with the somatic variant analysis, copy number profiling supported the clonal relationship between PR-259 and PR-285, and identified highly enriched, focal, high-level copy number alterations in the post-therapy NePC specimen.

Discussion

Small cell carcinoma/(NePC) is a rare PCa variant with an aggressive phenotype. Although de novo NePC constitutes <1 % of all PCa, autopsy series of castration-resistant prostate cancer (CRPCa) suggest the presence of NePC in 10–25 % of cases [8]. Despite high initial overall response rates (75–85 %) to platinum combinations, relapse to a chemo-refractory state is nearly universal with a median survival of less than 18 months [9].
Although initially posited as due to clonal selection of malignant neuroendocrine cells [10, 11], recent genetic evidence supports a model of NePC development due to transformation of prostate adenocarcinoma cells to a neuroendocrine phenotype, termed transdifferentiation (see review [12]). Consistent with the concept of a common clonal origin, recurrent prostate adenocarcinoma-specific alterations, such as recurrent ETS gene rearrangements, show concordant status in PCa admixed with NePC, and ETS rearrangement frequency is similar in conventional PCa and NePC [1315]. Additionally, identical mutations in the DNA-binding domains of TP53 have been observed in paired prostate adenocarcinoma and NePC [16]. The molecular mechanism of NePC development via transdifferentiation is also supported by a recent report showing gene amplification of AURKA and MYCN present in 65 % of adenocarcinomas that develop into NePC following ADT whereas only 5 % of unselected adenocarcinomas showing similar amplifications [17]. Lastly, RNAseq profiling in matched NePC and prostate adenocarcinomas showed downregulation of the transcriptional complex REST, which is integral to the repression of neuronal differentiation [18].
Despite the growing evidence in support of transdifferentiation, limited data has been published comparing comprehensive molecular profiling of a primary prostate adenocarcinoma and the subsequent metastatic NePC. Our current study reflects, to our knowledge, the first comprehensive profiling of paired diagnostic FFPE biopsy and subsequent NePC specimens. The presence of a somatic, deleterious SMAD4 variant in both the diagnostic and NePC specimens is consistent with clonal origin and the transdifferentiation model. Among 816 sequenced PCas in the cBioPortal database [19], six (0.7 %) harbor somatic SMAD4 mutations, including one sample with two SMAD4 mutations (Table 3). Importantly, six of the seven reported mutations impact the MH2 domain (including known inactivating and truncating mutations [20,21]) and are recurrent in the COSMIC database [22]. Of note, detailed mechanistic studies demonstrate that SMAD4 loss leads to an aggressive PCa phenotype in mouse models [23], providing a likely candidate driver of the aggressive phenotype in this exceptional “non-responder.”
Table 3
SMAD4 mutation and MYCL1 amplification frequency in prostate cancer NGS and copy number profiling studies available in cBioPortal
Study
Sample types
Cases with mutation data (n)
Cases with CNA data (n)
Cases with SMAD4 mutations (%)
Cases with MYCL amplifications (%)
Prostate (Broad/Cornell 2013)
PCa
57
56
1 (1.8 %)
0 (0 %)
Prostate (TCGA 2015)
PCa
333
492
3 (0.9 %)
0 (0 %)
Prostate (Broad/Cornell 2012)
PCa
112
109
1 (0.9 %)
0 (0 %)
Prostate (MSKCC 2014)
PCa
N/A
104
N/A
0 (0 %)
Prostate (MICH)
PCa and CRPC
61
61
1 (1.6 %)
0 (0 %)
Prostate (MSKCC 2010)
PCa and CRPC
103
194
0 (0 %)
0 (0 %)
Prostate (SU2C)
CRPC
150
150
0 (0 %)
0 (0 %)
Hovelson et al. 2015
PCa and CRPC
N/A*
116
N/A
1^ (0.8 %)
Total
 
816
1282
6 (0.7 %)
1 (0.08 %)
cBioPortal was queried for SMAD4 mutations and MYCL amplifications in prostate cancer tissue profiling studies. Sample types (localized untreated prostate adenocarcinoma [PCa] and/or castration-resistant prostate cancer [CRPC], the number of cases with mutation or copy number alteration (CNA) data, and the number (and %) of cases with SMAD4 mutations and MYCL amplifications are given
^,*Data from our recent targeted NGS study (Hovelson et al. 2015) which assessed MYCL but not SMAD4 is also included. The MYCL amplified case in that study was small cell/neuroendocrine prostate cancer (NePC). Totals from cBioPortal and our previous study for each parameter is given
The TP53 p.R282W mutation enrichment and homozygous RB1 loss in the NePC sample herein supports single gene studies and our recent targeted NGS profiling of eight NePC that show frequent inactivation of these genes in NePC [7, 24, 25]. Likewise, in a recent study, we used qRT-PCR and a combination of exome/targeted NGS to profile distinct conventional PCa and NePC components from an FFPE transurethral resection specimen, which demonstrated enrichment of a TP53 p.N151fs mutation exclusively in the NePC component [26]. Although both oncogenic and metastasis suppressive roles for MAP2K4 have been reported in PCa [2729], its role in NePC has not been described and will require additional investigation.
As described above, recurrent MYCN amplifications have been well-described in NePC [17]. Although a recent report identified recurrent MYCL amplifications in ~25 % of untreated Gleason score 7 PCa (>2 copies in 8–20 % of malignant glands) [30], clonal, high-level MYCL amplifications have not been observed in 1166 prior SNP-, aCGH-, or NGS-based copy number profiled untreated PCa or CRPC in cBioPortal (Table 3). However, in our previous NGS-based profiling of 116 aggressive PCas, we identified a single NePC (of 8 profiled) that harbored a high-level MCYL amplification (Additional file 2: Figure S1) [7]. Likewise, copy number profiling of mouse NePC resulting from prostate-specific p53 and RB inactivation identified recurrent MYCL gains [31]. MYCL amplifications and gene fusions have also been identified and shown to drive proliferation in small cell lung carcinomas [3234]. Taken together with our previous NGS profiling study, herein we identify recurrent MYCL1 amplifications in NePC, which will need to be confirmed in additional NePC cohorts.
Alternate mechanisms for the development and maintenance of NE transdifferentiation have been described. The process of “epithelial plasticity” provides evidence for the diverse phenotype of NE-like tumor cells, such as the variable expression of epithelial and NE markers following androgen deprivation [12, 3537]. This plasticity, which can occur via epithelial-to-mesenchymal transition (EMT) or mesenchymal-to-epithelial transition (MET), is regulated by a complex system of transcriptional networks and signaling pathways. The TMPRSS2-ERG fusion gene and certain microRNAs (i.e., miR-200 family) appear to promote the EMT phenotype leading, in part, to the castrate-resistant state [38, 39].
Importantly, neuroendocrine cancers involving other organ sites appear to have distinct molecular aberrations and highlight the need for individualized therapies [40]. For example, telomerase reverse transcriptase (TERT) promoter mutations are observed in many human epithelial cancers as well as the vast majority of urothelial neuroendocrine carcinomas, however, they are rarely found in NE-prostate or -lung cancers [41, 42]. Given the molecular heterogeneity of neuroendocrine carcinomas, targeted approaches guided by appropriate biomarker identification, rather than or in addition to cytotoxic therapies, are paramount to improve outcomes [43].
A variety of novel therapeutics targeting receptor tyrosine kinases, mammalian target of rapamycin (mTOR), angiogenesis, cell cycle, epigenetics, and immunotherapy have been tested, largely in small cell lung cancer, with limited success [44, 45]. Specific to the mutational landscape of NEPC, a number of targeted therapies have been investigated in in vitro and in murine models with varied success (see review [46]). Targeting tumor suppressor loss (TP53, RB1, and PTEN) is particularly relevant to NEPC given the high frequency of these alterations. For example, SAR405838, a novel small molecule inhibitor of the oncoprotein murine double minute 2 (MDM2)-TP53 protein-protein interaction, showed significant activity in wild-type TP53 murine models, including LNCaP prostate cancer lines [47]. Multiple small molecules that can activate TP53 are in early phase clinical trials, however, none at this time are recruiting patients with NEPC [46,48]. A phase II study of MLN8237, a small molecule inhibitor of Aurora Kinase A, is currently the only molecularly targeted trial enrolling men with CRPC with neuroendocrine features (NCT01799278).
A limitation of the current report is that it is based on NGS from one patient. Future case studies should consider application of immunohistochemical and morphoproteomic analyses, which might elucidate alternative mechanisms of resistance. The application of these tools has previously revealed means of response and resistance in two cases of refractory Ewing sarcoma that responded to combination therapy with insulin-like growth factor 1 receptor and mTOR inhibition [49].
“N of 1” cases provide unique hypothesis-generating opportunities with the potential to provide new information about pathogenic mechanisms and/or therapeutic response [2, 4]. We suggest that profiling of “exceptional non-responders” and temporally/histologically distinct tumor components [26, 50], as shown herein, may be as informative as “exceptional responder” studies and can exploit the wealth of archived diagnostic tissue specimens. Such studies may be particularly important for identifying the prognostic and predictive associations of rare alterations, such as SMAD4 mutations in prostate cancer, as well as identifying adaptive alterations associated with treatment resistance/progression such as MCYL amplifications.

Conclusions

Through comprehensive profiling of archived diagnostic and liver biopsy specimens from a single patient with an aggressive clinical course, we identify molecular alterations associated with rapid progression from prostatic adenocarcinoma to NePC, and more broadly identify MYCL as a recurrently amplified gene specifically in NePC.

Funding/Support

This work was supported in part by the Evans Foundation/Prostate Cancer Foundation (to S.A.T), the National Institutes of Health (R01 CA183857 to S.A.T) and the Department of Defense (PC120464 to K.A.C.). S.A.T. was supported by University of Michigan Prostate SPORE Career Development Award and the A. Alfred Taubman Medical Research Institute.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Competing interests

The University of Michigan has been issued a patent on the detection of ETS gene fusions in prostate cancer, on which S.A.T is a co-inventor. The University of Michigan licensed the diagnostic field of use to Gen-Probe, Inc., who has sublicensed some rights to Ventana/Roche. S.A.T. serves as a consultant to, and has received honoraria from, Ventana/Roche. No other disclosures are reported.

Authors’ contributions

KK and ST contributed equally to the study. ST participated in the conception, design, funding, study supervision, and carried out the molecular genetic studies, participated in the analysis and interpretation of the data as well as drafting, and critical review of the manuscript. KK participated in the conception, design, analysis, and interpretation of the data, as well as drafting and critical review of the manuscript. SS participated in the analysis and interpretation of the data and drafting of the manuscript. AC, KO, DH, CL all participated in the acquisition, analysis, and interpretation of the data as well as the administrative, technical, and material support. DH also performed the statistical analysis. KC participated in the conception, design, funding, and study supervision, and participated in the analysis and interpretation of the data as well as drafting and critical review of the manuscript. All authors read and approved the final manuscript.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Al-Ahmadie H, Iyer G, Hohl M, Asthana S, Inagaki A, Schultz N, et al. Synthetic lethality in ATM-deficient RAD50-mutant tumors underlies outlier response to cancer therapy. Cancer Discov. 2014;4(9):1014–21.PubMedCentralCrossRefPubMed Al-Ahmadie H, Iyer G, Hohl M, Asthana S, Inagaki A, Schultz N, et al. Synthetic lethality in ATM-deficient RAD50-mutant tumors underlies outlier response to cancer therapy. Cancer Discov. 2014;4(9):1014–21.PubMedCentralCrossRefPubMed
3.
Zurück zum Zitat Iyer G, Hanrahan AJ, Milowsky MI, Al-Ahmadie H, Scott SN, Janakiraman M, et al. Genome sequencing identifies a basis for everolimus sensitivity. Science. 2012;338(6104):221.PubMedCentralCrossRefPubMed Iyer G, Hanrahan AJ, Milowsky MI, Al-Ahmadie H, Scott SN, Janakiraman M, et al. Genome sequencing identifies a basis for everolimus sensitivity. Science. 2012;338(6104):221.PubMedCentralCrossRefPubMed
4.
Zurück zum Zitat Subbiah IM, Subbiah V. Exceptional responders: in search of the science behind the miracle cancer cures. Future Oncol. 2015;11(1):1–4.CrossRefPubMed Subbiah IM, Subbiah V. Exceptional responders: in search of the science behind the miracle cancer cures. Future Oncol. 2015;11(1):1–4.CrossRefPubMed
5.
Zurück zum Zitat Grasso C, Butler T, Rhodes K, Quist M, Neff TL, Moore S, et al. Assessing copy number alterations in targeted, amplicon-based next-generation sequencing data. J Mol Diagn. 2015;17(1):53–63.CrossRefPubMed Grasso C, Butler T, Rhodes K, Quist M, Neff TL, Moore S, et al. Assessing copy number alterations in targeted, amplicon-based next-generation sequencing data. J Mol Diagn. 2015;17(1):53–63.CrossRefPubMed
6.
Zurück zum Zitat Warrick JI, Hovelson DH, Amin A, Liu CJ, Cani AK, McDaniel AS, et al. Tumor evolution and progression in multifocal and paired non-invasive/invasive urothelial carcinoma. Virchows Arch. 2015;466(3):297–311.CrossRefPubMed Warrick JI, Hovelson DH, Amin A, Liu CJ, Cani AK, McDaniel AS, et al. Tumor evolution and progression in multifocal and paired non-invasive/invasive urothelial carcinoma. Virchows Arch. 2015;466(3):297–311.CrossRefPubMed
7.
Zurück zum Zitat Hovelson DH, McDaniel AS, Cani AK, Johnson B, Rhodes K, Williams PD, et al. Development and validation of a scalable next-generation sequencing system for assessing relevant somatic variants in solid tumors. Neoplasia. 2015;17(4):385–99.PubMedCentralCrossRefPubMed Hovelson DH, McDaniel AS, Cani AK, Johnson B, Rhodes K, Williams PD, et al. Development and validation of a scalable next-generation sequencing system for assessing relevant somatic variants in solid tumors. Neoplasia. 2015;17(4):385–99.PubMedCentralCrossRefPubMed
8.
Zurück zum Zitat Shah RB, Mehra R, Chinnaiyan AM, Shen R, Ghosh D, Zhou M, et al. Androgen-independent prostate cancer is a heterogeneous group of diseases: lessons from a rapid autopsy program. Cancer Res. 2004;64(24):9209–16.CrossRefPubMed Shah RB, Mehra R, Chinnaiyan AM, Shen R, Ghosh D, Zhou M, et al. Androgen-independent prostate cancer is a heterogeneous group of diseases: lessons from a rapid autopsy program. Cancer Res. 2004;64(24):9209–16.CrossRefPubMed
9.
Zurück zum Zitat Deorah S, Rao MB, Raman R, Gaitonde K, Donovan JF. Survival of patients with small cell carcinoma of the prostate during 1973–2003: a population-based study. BJU Int. 2012;109(6):824–30.CrossRefPubMed Deorah S, Rao MB, Raman R, Gaitonde K, Donovan JF. Survival of patients with small cell carcinoma of the prostate during 1973–2003: a population-based study. BJU Int. 2012;109(6):824–30.CrossRefPubMed
10.
Zurück zum Zitat Yashi M, Terauchi F, Nukui A, Ochi M, Yuzawa M, Hara Y, et al. Small-cell neuroendocrine carcinoma as a variant form of prostate cancer recurrence: a case report and short literature review. Urol Oncol. 2006;24(4):313–7.CrossRefPubMed Yashi M, Terauchi F, Nukui A, Ochi M, Yuzawa M, Hara Y, et al. Small-cell neuroendocrine carcinoma as a variant form of prostate cancer recurrence: a case report and short literature review. Urol Oncol. 2006;24(4):313–7.CrossRefPubMed
11.
Zurück zum Zitat Tanaka M, Suzuki Y, Takaoka K, Suzuki N, Murakami S, Matsuzaki O, et al. Progression of prostate cancer to neuroendocrine cell tumor. Int J Urol. 2001;8(8):431–6. discussion 437.CrossRefPubMed Tanaka M, Suzuki Y, Takaoka K, Suzuki N, Murakami S, Matsuzaki O, et al. Progression of prostate cancer to neuroendocrine cell tumor. Int J Urol. 2001;8(8):431–6. discussion 437.CrossRefPubMed
13.
Zurück zum Zitat Williamson SR, Zhang S, Yao JL, Huang J, Lopez-Beltran A, Shen S, et al. ERG-TMPRSS2 rearrangement is shared by concurrent prostatic adenocarcinoma and prostatic small cell carcinoma and absent in small cell carcinoma of the urinary bladder: evidence supporting monoclonal origin. Mod Pathol. 2011;24(8):1120–7.PubMedCentralCrossRefPubMed Williamson SR, Zhang S, Yao JL, Huang J, Lopez-Beltran A, Shen S, et al. ERG-TMPRSS2 rearrangement is shared by concurrent prostatic adenocarcinoma and prostatic small cell carcinoma and absent in small cell carcinoma of the urinary bladder: evidence supporting monoclonal origin. Mod Pathol. 2011;24(8):1120–7.PubMedCentralCrossRefPubMed
14.
Zurück zum Zitat Han B, Mehra R, Suleman K, Tomlins SA, Wang L, Singhal N, et al. Characterization of ETS gene aberrations in select histologic variants of prostate carcinoma. Mod Pathol. 2009;22(9):1176–85.PubMedCentralCrossRefPubMed Han B, Mehra R, Suleman K, Tomlins SA, Wang L, Singhal N, et al. Characterization of ETS gene aberrations in select histologic variants of prostate carcinoma. Mod Pathol. 2009;22(9):1176–85.PubMedCentralCrossRefPubMed
15.
Zurück zum Zitat Lotan TL, Gupta NS, Wang W, Toubaji A, Haffner MC, Chaux A, et al. ERG gene rearrangements are common in prostatic small cell carcinomas. Mod Pathol. 2011;24(6):820–8.PubMedCentralCrossRefPubMed Lotan TL, Gupta NS, Wang W, Toubaji A, Haffner MC, Chaux A, et al. ERG gene rearrangements are common in prostatic small cell carcinomas. Mod Pathol. 2011;24(6):820–8.PubMedCentralCrossRefPubMed
16.
Zurück zum Zitat Hansel DE, Nakayama M, Luo J, Abukhdeir AM, Park BH, Bieberich CJ, et al. Shared TP53 gene mutation in morphologically and phenotypically distinct concurrent primary small cell neuroendocrine carcinoma and adenocarcinoma of the prostate. Prostate. 2009;69(6):603–9.PubMedCentralCrossRefPubMed Hansel DE, Nakayama M, Luo J, Abukhdeir AM, Park BH, Bieberich CJ, et al. Shared TP53 gene mutation in morphologically and phenotypically distinct concurrent primary small cell neuroendocrine carcinoma and adenocarcinoma of the prostate. Prostate. 2009;69(6):603–9.PubMedCentralCrossRefPubMed
17.
Zurück zum Zitat Beltran H, Rickman DS, Park K, Chae SS, Sboner A, MacDonald TY, et al. Molecular characterization of neuroendocrine prostate cancer and identification of new drug targets. Cancer Discov. 2011;1(6):487–95.PubMedCentralCrossRefPubMed Beltran H, Rickman DS, Park K, Chae SS, Sboner A, MacDonald TY, et al. Molecular characterization of neuroendocrine prostate cancer and identification of new drug targets. Cancer Discov. 2011;1(6):487–95.PubMedCentralCrossRefPubMed
18.
Zurück zum Zitat Lapuk AV, Wu C, Wyatt AW, McPherson A, McConeghy BJ, Brahmbhatt S, et al. From sequence to molecular pathology, and a mechanism driving the neuroendocrine phenotype in prostate cancer. J Pathol. 2012;227(3):286–97.PubMedCentralCrossRefPubMed Lapuk AV, Wu C, Wyatt AW, McPherson A, McConeghy BJ, Brahmbhatt S, et al. From sequence to molecular pathology, and a mechanism driving the neuroendocrine phenotype in prostate cancer. J Pathol. 2012;227(3):286–97.PubMedCentralCrossRefPubMed
19.
Zurück zum Zitat Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012;2(5):401–4.CrossRefPubMed Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012;2(5):401–4.CrossRefPubMed
20.
Zurück zum Zitat De Bosscher K, Hill CS, Nicolas FJ. Molecular and functional consequences of Smad4 C-terminal missense mutations in colorectal tumour cells. Biochem J. 2004;379(Pt 1):209–16.PubMedCentralCrossRefPubMed De Bosscher K, Hill CS, Nicolas FJ. Molecular and functional consequences of Smad4 C-terminal missense mutations in colorectal tumour cells. Biochem J. 2004;379(Pt 1):209–16.PubMedCentralCrossRefPubMed
21.
Zurück zum Zitat Maurice D, Pierreux CE, Howell M, Wilentz RE, Owen MJ, Hill CS. Loss of Smad4 function in pancreatic tumors: C-terminal truncation leads to decreased stability. J Biol Chem. 2001;276(46):43175–81.CrossRefPubMed Maurice D, Pierreux CE, Howell M, Wilentz RE, Owen MJ, Hill CS. Loss of Smad4 function in pancreatic tumors: C-terminal truncation leads to decreased stability. J Biol Chem. 2001;276(46):43175–81.CrossRefPubMed
22.
Zurück zum Zitat Forbes SA, Beare D, Gunasekaran P, Leung K, Bindal N, Boutselakis H, et al. COSMIC: exploring the world's knowledge of somatic mutations in human cancer. Nucleic Acids Res. 2015;43(Database issue):D805–11.PubMedCentralCrossRefPubMed Forbes SA, Beare D, Gunasekaran P, Leung K, Bindal N, Boutselakis H, et al. COSMIC: exploring the world's knowledge of somatic mutations in human cancer. Nucleic Acids Res. 2015;43(Database issue):D805–11.PubMedCentralCrossRefPubMed
23.
Zurück zum Zitat Ding Z, Wu CJ, Chu GC, Xiao Y, Ho D, Zhang J, et al. SMAD4-dependent barrier constrains prostate cancer growth and metastatic progression. Nature. 2011;470(7333):269–73.PubMedCentralCrossRefPubMed Ding Z, Wu CJ, Chu GC, Xiao Y, Ho D, Zhang J, et al. SMAD4-dependent barrier constrains prostate cancer growth and metastatic progression. Nature. 2011;470(7333):269–73.PubMedCentralCrossRefPubMed
24.
Zurück zum Zitat Tan HL, Sood A, Rahimi HA, Wang W, Gupta N, Hicks J, et al. Rb loss is characteristic of prostatic small cell neuroendocrine carcinoma. Clin Cancer Res. 2014;20(4):890–903.PubMedCentralCrossRefPubMed Tan HL, Sood A, Rahimi HA, Wang W, Gupta N, Hicks J, et al. Rb loss is characteristic of prostatic small cell neuroendocrine carcinoma. Clin Cancer Res. 2014;20(4):890–903.PubMedCentralCrossRefPubMed
25.
Zurück zum Zitat Chen H, Sun Y, Wu C, Magyar CE, Li X, Cheng L, et al. Pathogenesis of prostatic small cell carcinoma involves the inactivation of the P53 pathway. Endocr Relat Cancer. 2012;19(3):321–31.PubMedCentralCrossRefPubMed Chen H, Sun Y, Wu C, Magyar CE, Li X, Cheng L, et al. Pathogenesis of prostatic small cell carcinoma involves the inactivation of the P53 pathway. Endocr Relat Cancer. 2012;19(3):321–31.PubMedCentralCrossRefPubMed
26.
Zurück zum Zitat Grasso CS, Cani AK, Hovelson DH, Quist MJ, Douville NJ, Yadati V, et al. Integrative molecular profiling of routine clinical prostate cancer specimens. Ann Oncol. 2015;26(6):1110–8.CrossRefPubMed Grasso CS, Cani AK, Hovelson DH, Quist MJ, Douville NJ, Yadati V, et al. Integrative molecular profiling of routine clinical prostate cancer specimens. Ann Oncol. 2015;26(6):1110–8.CrossRefPubMed
27.
Zurück zum Zitat Pavese JM, Ogden IM, Voll EA, Huang X, Xu L, Jovanovic B, et al. Mitogen-activated protein kinase kinase 4 (MAP2K4) promotes human prostate cancer metastasis. PLoS One. 2014;9(7):e102289.PubMedCentralCrossRefPubMed Pavese JM, Ogden IM, Voll EA, Huang X, Xu L, Jovanovic B, et al. Mitogen-activated protein kinase kinase 4 (MAP2K4) promotes human prostate cancer metastasis. PLoS One. 2014;9(7):e102289.PubMedCentralCrossRefPubMed
28.
Zurück zum Zitat Kim HL, Vander Griend DJ, Yang X, Benson DA, Dubauskas Z, Yoshida BA, et al. Mitogen-activated protein kinase kinase 4 metastasis suppressor gene expression is inversely related to histological pattern in advancing human prostatic cancers. Cancer Res. 2001;61(7):2833–7.PubMed Kim HL, Vander Griend DJ, Yang X, Benson DA, Dubauskas Z, Yoshida BA, et al. Mitogen-activated protein kinase kinase 4 metastasis suppressor gene expression is inversely related to histological pattern in advancing human prostatic cancers. Cancer Res. 2001;61(7):2833–7.PubMed
29.
Zurück zum Zitat Yoshida BA, Dubauskas Z, Chekmareva MA, Christiano TR, Stadler WM, Rinker-Schaeffer CW. Mitogen-activated protein kinase kinase 4/stress-activated protein/Erk kinase 1 (MKK4/SEK1), a prostate cancer metastasis suppressor gene encoded by human chromosome 17. Cancer Res. 1999;59(21):5483–7.PubMed Yoshida BA, Dubauskas Z, Chekmareva MA, Christiano TR, Stadler WM, Rinker-Schaeffer CW. Mitogen-activated protein kinase kinase 4/stress-activated protein/Erk kinase 1 (MKK4/SEK1), a prostate cancer metastasis suppressor gene encoded by human chromosome 17. Cancer Res. 1999;59(21):5483–7.PubMed
30.
Zurück zum Zitat Boutros PC, Fraser M, Harding NJ, de Borja R, Trudel D, Lalonde E, et al. Spatial genomic heterogeneity within localized, multifocal prostate cancer. Nat Genet. 2015;47(7):736–45.CrossRefPubMed Boutros PC, Fraser M, Harding NJ, de Borja R, Trudel D, Lalonde E, et al. Spatial genomic heterogeneity within localized, multifocal prostate cancer. Nat Genet. 2015;47(7):736–45.CrossRefPubMed
31.
Zurück zum Zitat Zhou Z, Flesken-Nikitin A, Corney DC, Wang W, Goodrich DW, Roy-Burman P, et al. Synergy of p53 and Rb deficiency in a conditional mouse model for metastatic prostate cancer. Cancer Res. 2006;66(16):7889–98.CrossRefPubMed Zhou Z, Flesken-Nikitin A, Corney DC, Wang W, Goodrich DW, Roy-Burman P, et al. Synergy of p53 and Rb deficiency in a conditional mouse model for metastatic prostate cancer. Cancer Res. 2006;66(16):7889–98.CrossRefPubMed
32.
Zurück zum Zitat Peifer M, Fernandez-Cuesta L, Sos ML, George J, Seidel D, Kasper LH, et al. Integrative genome analyses identify key somatic driver mutations of small-cell lung cancer. Nat Genet. 2012;44(10):1104–10.CrossRefPubMed Peifer M, Fernandez-Cuesta L, Sos ML, George J, Seidel D, Kasper LH, et al. Integrative genome analyses identify key somatic driver mutations of small-cell lung cancer. Nat Genet. 2012;44(10):1104–10.CrossRefPubMed
33.
Zurück zum Zitat Rudin CM, Durinck S, Stawiski EW, Poirier JT, Modrusan Z, Shames DS, et al. Comprehensive genomic analysis identifies SOX2 as a frequently amplified gene in small-cell lung cancer. Nat Genet. 2012;44(10):1111–6.PubMedCentralCrossRefPubMed Rudin CM, Durinck S, Stawiski EW, Poirier JT, Modrusan Z, Shames DS, et al. Comprehensive genomic analysis identifies SOX2 as a frequently amplified gene in small-cell lung cancer. Nat Genet. 2012;44(10):1111–6.PubMedCentralCrossRefPubMed
34.
Zurück zum Zitat Varghese AM, Zakowski MF, Yu HA, Won HH, Riely GJ, Krug LM, et al. Small-cell lung cancers in patients who never smoked cigarettes. J Thorac Oncol. 2014;9(6):892–6.PubMedCentralCrossRefPubMed Varghese AM, Zakowski MF, Yu HA, Won HH, Riely GJ, Krug LM, et al. Small-cell lung cancers in patients who never smoked cigarettes. J Thorac Oncol. 2014;9(6):892–6.PubMedCentralCrossRefPubMed
35.
Zurück zum Zitat Nieto MA. Epithelial plasticity: a common theme in embryonic and cancer cells. Science. 2013;342(6159):1234850.CrossRefPubMed Nieto MA. Epithelial plasticity: a common theme in embryonic and cancer cells. Science. 2013;342(6159):1234850.CrossRefPubMed
36.
Zurück zum Zitat Nordin A, Wang W, Welen K, Damber JE. Midkine is associated with neuroendocrine differentiation in castration-resistant prostate cancer. Prostate. 2013;73(6):657–67.CrossRefPubMed Nordin A, Wang W, Welen K, Damber JE. Midkine is associated with neuroendocrine differentiation in castration-resistant prostate cancer. Prostate. 2013;73(6):657–67.CrossRefPubMed
37.
Zurück zum Zitat Terry S, Maille P, Baaddi H, Kheuang L, Soyeux P, Nicolaiew N, et al. Cross modulation between the androgen receptor axis and protocadherin-PC in mediating neuroendocrine transdifferentiation and therapeutic resistance of prostate cancer. Neoplasia. 2013;15(7):761–72.PubMedCentralCrossRefPubMed Terry S, Maille P, Baaddi H, Kheuang L, Soyeux P, Nicolaiew N, et al. Cross modulation between the androgen receptor axis and protocadherin-PC in mediating neuroendocrine transdifferentiation and therapeutic resistance of prostate cancer. Neoplasia. 2013;15(7):761–72.PubMedCentralCrossRefPubMed
38.
Zurück zum Zitat Cerasuolo M, Paris D, Iannotti FA, Melck D, Verde R, Mazzarella E, et al. Neuroendocrine transdifferentiation in human prostate cancer cells: an integrated approach. Cancer Res. 2015;75(15):2975–86.CrossRefPubMed Cerasuolo M, Paris D, Iannotti FA, Melck D, Verde R, Mazzarella E, et al. Neuroendocrine transdifferentiation in human prostate cancer cells: an integrated approach. Cancer Res. 2015;75(15):2975–86.CrossRefPubMed
39.
Zurück zum Zitat Leshem O, Madar S, Kogan-Sakin I, Kamer I, Goldstein I, Brosh R, et al. TMPRSS2/ERG promotes epithelial to mesenchymal transition through the ZEB1/ZEB2 axis in a prostate cancer model. PLoS One. 2011;6(7):e21650.PubMedCentralCrossRefPubMed Leshem O, Madar S, Kogan-Sakin I, Kamer I, Goldstein I, Brosh R, et al. TMPRSS2/ERG promotes epithelial to mesenchymal transition through the ZEB1/ZEB2 axis in a prostate cancer model. PLoS One. 2011;6(7):e21650.PubMedCentralCrossRefPubMed
40.
Zurück zum Zitat Zheng X, Liu D, Fallon JT, Zhong M. Distinct genetic alterations in small cell carcinoma from different anatomic sites. Exp Hematol Oncol. 2015;4:2.PubMedCentralCrossRefPubMed Zheng X, Liu D, Fallon JT, Zhong M. Distinct genetic alterations in small cell carcinoma from different anatomic sites. Exp Hematol Oncol. 2015;4:2.PubMedCentralCrossRefPubMed
41.
Zurück zum Zitat Stoehr R, Taubert H, Zinnall U, Giedl J, Gaisa NT, Burger M, et al. Frequency of TERT promoter mutations in prostate cancer. Pathobiology. 2015;82(2):53–7.CrossRefPubMed Stoehr R, Taubert H, Zinnall U, Giedl J, Gaisa NT, Burger M, et al. Frequency of TERT promoter mutations in prostate cancer. Pathobiology. 2015;82(2):53–7.CrossRefPubMed
42.
Zurück zum Zitat Zheng X, Zhuge J, Bezerra SM, Faraj SF, Munari E, Fallon JT, 3rd, et al. High frequency of TERT promoter mutation in small cell carcinoma of bladder, but not in small cell carcinoma of other origins. J Hematol Oncol. 2014;7:47. Zheng X, Zhuge J, Bezerra SM, Faraj SF, Munari E, Fallon JT, 3rd, et al. High frequency of TERT promoter mutation in small cell carcinoma of bladder, but not in small cell carcinoma of other origins. J Hematol Oncol. 2014;7:47.
44.
Zurück zum Zitat Arcaro A. Targeted therapies for small cell lung cancer: Where do we stand? Crit Rev Oncol Hematol. 2015;95(2):154–64.CrossRefPubMed Arcaro A. Targeted therapies for small cell lung cancer: Where do we stand? Crit Rev Oncol Hematol. 2015;95(2):154–64.CrossRefPubMed
45.
Zurück zum Zitat Ehrlich D, Wang B, Lu W, Dowling P, Yuan R. Intratumoral anti-HuD immunotoxin therapy for small cell lung cancer and neuroblastoma. J Hematol Oncol. 2014;7(1):91.PubMedCentralCrossRefPubMed Ehrlich D, Wang B, Lu W, Dowling P, Yuan R. Intratumoral anti-HuD immunotoxin therapy for small cell lung cancer and neuroblastoma. J Hematol Oncol. 2014;7(1):91.PubMedCentralCrossRefPubMed
46.
47.
Zurück zum Zitat Wang S, Sun W, Zhao Y, McEachern D, Meaux I, Barriere C, et al. SAR405838: an optimized inhibitor of MDM2-p53 interaction that induces complete and durable tumor regression. Cancer Res. 2014;74(20):5855–65.CrossRefPubMed Wang S, Sun W, Zhao Y, McEachern D, Meaux I, Barriere C, et al. SAR405838: an optimized inhibitor of MDM2-p53 interaction that induces complete and durable tumor regression. Cancer Res. 2014;74(20):5855–65.CrossRefPubMed
49.
Zurück zum Zitat Subbiah V, Naing A, Brown RE, Chen H, Doyle L, LoRusso P, et al. Targeted morphoproteomic profiling of Ewing's sarcoma treated with insulin-like growth factor 1 receptor (IGF1R) inhibitors: response/resistance signatures. PLoS One. 2011;6(4):e18424.PubMedCentralCrossRefPubMed Subbiah V, Naing A, Brown RE, Chen H, Doyle L, LoRusso P, et al. Targeted morphoproteomic profiling of Ewing's sarcoma treated with insulin-like growth factor 1 receptor (IGF1R) inhibitors: response/resistance signatures. PLoS One. 2011;6(4):e18424.PubMedCentralCrossRefPubMed
50.
Zurück zum Zitat Haffner MC, Mosbruger T, Esopi DM, Fedor H, Heaphy CM, Walker DA, et al. Tracking the clonal origin of lethal prostate cancer. J Clin Invest. 2013;123(11):4918–22.PubMedCentralCrossRefPubMed Haffner MC, Mosbruger T, Esopi DM, Fedor H, Heaphy CM, Walker DA, et al. Tracking the clonal origin of lethal prostate cancer. J Clin Invest. 2013;123(11):4918–22.PubMedCentralCrossRefPubMed
Metadaten
Titel
Comprehensive serial molecular profiling of an “N of 1” exceptional non-responder with metastatic prostate cancer progressing to small cell carcinoma on treatment
verfasst von
Kunal C. Kadakia
Scott A. Tomlins
Saagar K. Sanghvi
Andi K. Cani
Kei Omata
Daniel H. Hovelson
Chia-Jen Liu
Kathleen A. Cooney
Publikationsdatum
01.12.2015
Verlag
BioMed Central
Erschienen in
Journal of Hematology & Oncology / Ausgabe 1/2015
Elektronische ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-015-0204-7

Weitere Artikel der Ausgabe 1/2015

Journal of Hematology & Oncology 1/2015 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.