Network


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

Hotspot


Dive into the research topics where Matthias Holdhoff is active.

Publication


Featured researches published by Matthias Holdhoff.


Science Translational Medicine | 2014

Detection of Circulating Tumor DNA in Early- and Late-Stage Human Malignancies

Chetan Bettegowda; Mark Sausen; Rebecca J. Leary; Isaac Kinde; Yuxuan Wang; Nishant Agrawal; Bjarne Bartlett; Hao Wang; Brandon Luber; Rhoda M. Alani; Emmanuel S. Antonarakis; Nilofer Saba Azad; Alberto Bardelli; Henry Brem; John L. Cameron; Clarence Lee; Leslie A. Fecher; Gary L. Gallia; Peter Gibbs; Dung Le; Robert L. Giuntoli; Michael Goggins; Michael D. Hogarty; Matthias Holdhoff; Seung-Mo Hong; Yuchen Jiao; Hartmut H. Juhl; Jenny J. Kim; Giulia Siravegna; Daniel A. Laheru

Circulating tumor DNA can be used in a variety of clinical and investigational settings across tumor types and stages for screening, diagnosis, and identifying mutations responsible for therapeutic response and drug resistance. Circulating Tumor DNA for Early Detection and Managing Resistance Cancer evolves over time, without any warning signs. Similarly, the development of resistance to therapy generally becomes apparent only when there are obvious signs of tumor growth, at which point the patient may have lost valuable time. Although a repeat biopsy may be able to identify drug-resistant mutations before the tumor has a chance to regrow, it is usually not feasible to do many repeat biopsies. Now, two studies are demonstrating the utility of monitoring the patients’ blood for tumor DNA to detect cancer at the earliest stages of growth or resistance. In one study, Bettegowda and coauthors showed that sampling a patient’s blood may be sufficient to yield information about the tumor’s genetic makeup, even for many early-stage cancers, without a need for an invasive procedure to collect tumor tissue, such as surgery or endoscopy. The authors demonstrated the presence of circulating DNA from many types of tumors that had not yet metastasized or released detectable cells into the circulation. They could detect more than 50% of patients across 14 tumor types at the earliest stages, when these cancers may still be curable, suggesting that a blood draw could be a viable screening approach to detecting most cancers. They also showed that in patients with colorectal cancer, the information derived from circulating tumor DNA could be used to determine the optimal course of treatment and identify resistance to epidermal growth factor receptor (EGFR) blockade. Meanwhile, Misale and colleagues illustrated a way to use this information to overcome treatment resistance. These authors also found that mutations associated with EGFR inhibitor resistance could be detected in the blood of patients with colorectal cancer. In addition, they demonstrated that adding MEK inhibitors, another class of anticancer drugs, can successfully overcome resistance when given in conjunction with the EGFR inhibitors. Thus, the studies from Bettegowda and Misale and their colleagues show the effectiveness of analyzing circulating DNA from a variety of tumors and highlight the potential investigational and clinical applications of this novel technology for early detection, monitoring resistance, and devising treatment plans to overcome resistance. The development of noninvasive methods to detect and monitor tumors continues to be a major challenge in oncology. We used digital polymerase chain reaction–based technologies to evaluate the ability of circulating tumor DNA (ctDNA) to detect tumors in 640 patients with various cancer types. We found that ctDNA was detectable in >75% of patients with advanced pancreatic, ovarian, colorectal, bladder, gastroesophageal, breast, melanoma, hepatocellular, and head and neck cancers, but in less than 50% of primary brain, renal, prostate, or thyroid cancers. In patients with localized tumors, ctDNA was detected in 73, 57, 48, and 50% of patients with colorectal cancer, gastroesophageal cancer, pancreatic cancer, and breast adenocarcinoma, respectively. ctDNA was often present in patients without detectable circulating tumor cells, suggesting that these two biomarkers are distinct entities. In a separate panel of 206 patients with metastatic colorectal cancers, we showed that the sensitivity of ctDNA for detection of clinically relevant KRAS gene mutations was 87.2% and its specificity was 99.2%. Finally, we assessed whether ctDNA could provide clues into the mechanisms underlying resistance to epidermal growth factor receptor blockade in 24 patients who objectively responded to therapy but subsequently relapsed. Twenty-three (96%) of these patients developed one or more mutations in genes involved in the mitogen-activated protein kinase pathway. Together, these data suggest that ctDNA is a broadly applicable, sensitive, and specific biomarker that can be used for a variety of clinical and research purposes in patients with multiple different types of cancer.


Science | 2017

Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade

Dung T. Le; Jennifer N. Durham; Kellie Nicole Smith; Hao Wang; Bjarne Bartlett; Laveet K. Aulakh; Steve Lu; Holly Kemberling; Cara Wilt; Brandon Luber; Fay Wong; Nilofer Saba Azad; Agnieszka A. Rucki; Daniel A. Laheru; Ross C. Donehower; Atif Zaheer; George A. Fisher; Todd S. Crocenzi; James J. Lee; Tim F. Greten; Austin Duffy; Kristen K. Ciombor; Aleksandra Eyring; Bao H. Lam; Andrew K. Joe; S. Peter Kang; Matthias Holdhoff; Ludmila Danilova; Leslie Cope; Christian Meyer

Predicting responses to immunotherapy Colon cancers with loss-of-function mutations in the mismatch repair (MMR) pathway have favorable responses to PD-1 blockade immunotherapy. In a phase 2 clinical trial, Le et al. showed that treatment success is not just limited to colon cancer (see the Perspective by Goswami and Sharma). They found that a wide range of different cancer types with MMR deficiency also responded to PD-1 blockade. The trial included some patients with pancreatic cancer, which is one of the deadliest forms of cancer. The clinical trial is still ongoing, and around 20% of patients have so far achieved a complete response. MMR deficiency appears to be a biomarker for predicting successful treatment outcomes for several solid tumors and indicates a new therapeutic option for patients harboring MMR-deficient cancers. Science, this issue p. 409; see also p. 358 A pan-cancer biomarker is identified that can predict successful response to cancer immunotherapy in human patients. The genomes of cancers deficient in mismatch repair contain exceptionally high numbers of somatic mutations. In a proof-of-concept study, we previously showed that colorectal cancers with mismatch repair deficiency were sensitive to immune checkpoint blockade with antibodies to programmed death receptor–1 (PD-1). We have now expanded this study to evaluate the efficacy of PD-1 blockade in patients with advanced mismatch repair–deficient cancers across 12 different tumor types. Objective radiographic responses were observed in 53% of patients, and complete responses were achieved in 21% of patients. Responses were durable, with median progression-free survival and overall survival still not reached. Functional analysis in a responding patient demonstrated rapid in vivo expansion of neoantigen-specific T cell clones that were reactive to mutant neopeptides found in the tumor. These data support the hypothesis that the large proportion of mutant neoantigens in mismatch repair–deficient cancers make them sensitive to immune checkpoint blockade, regardless of the cancers’ tissue of origin.


Clinical Cancer Research | 2015

First-in-human dose study of the novel transforming growth factor-β receptor I kinase inhibitor LY2157299 monohydrate in patients with advanced cancer and glioma.

Jordi Rodon; Michael A. Carducci; Juan M. Sepulveda-Sánchez; Analia Azaro; Emiliano Calvo; Joan Seoane; Irene Braña; Elisabet Sicart; Ivelina Gueorguieva; Ann Cleverly; N. Sokalingum Pillay; Durisala Desaiah; Shawn T. Estrem; Luis Paz-Ares; Matthias Holdhoff; Jaishri O. Blakeley; Michael Lahn; José Baselga

Purpose: TGFβ signaling plays a key role in tumor progression, including malignant glioma. Small-molecule inhibitors such as LY2157299 monohydrate (LY2157299) block TGFβ signaling and reduce tumor progression in preclinical models. To use LY2157299 in the treatment of malignancies, we investigated its properties in a first-in-human dose (FHD) study in patients with cancer. Experimental Design: Sixty-five patients (58 with glioma) with measurable and progressive malignancies were enrolled. Oral LY2157299 was given as a split dose morning and evening on an intermittent schedule of 14 days on and 14 days off (28-day cycle). LY2157299 monotherapy was studied in dose escalation (part A) first and then evaluated in combination with standard doses of lomustine (part B). Safety was assessed using Common Terminology Criteria for Adverse Events version 3.0, echocardiography/Doppler imaging, serum troponin I, and brain natriuretic peptide (BNP) levels. Antitumor activity was assessed by RECIST and Macdonald criteria. Results: In part A, 16.6% (5/30) and in part B, 7.7% (2/26) of evaluable patients with glioma had either a complete (CR) or a partial response (PR). In both parts, 15 patients with glioma had stable disease (SD), 5 of whom had SD ≥6 cycles of treatment. Therefore, clinical benefit (CR+PR+SD ≥6 cycles) was observed in 12 of 56 patients with glioma (21.4%). LY2157299 was safe, with no cardiac adverse events. Conclusions: On the basis of the safety, pharmacokinetics, and antitumor activity in patients with glioma, the intermittent administration of LY2157299 at 300 mg/day is safe for future clinical investigation. Clin Cancer Res; 21(3); 553–60. ©2014 AACR.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Detection of tumor-derived DNA in cerebrospinal fluid of patients with primary tumors of the brain and spinal cord

Yuxuan Wang; Simeon Springer; Ming Zhang; K. Wyatt McMahon; Isaac Kinde; Lisa Dobbyn; Janine Ptak; Henry Brem; Kaisorn L. Chaichana; Gary L. Gallia; Ziya L. Gokaslan; Mari L. Groves; George I. Jallo; Michael Lim; Alessandro Olivi; Alfredo Quiñones-Hinojosa; Daniele Rigamonti; Greg Riggins; Daniel M. Sciubba; Jon D. Weingart; Jean Paul Wolinsky; Xiaobu Ye; Sueli Mieko Oba-Shinjo; Suely Kazue Nagahashi Marie; Matthias Holdhoff; Nishant Agrawal; Luis A. Diaz; Nickolas Papadopoulos; Kenneth W. Kinzler; Bert Vogelstein

Significance Outcomes for individuals with central nervous system (CNS) malignancies remain abysmal. A major challenge in managing these patients is the lack of reliable biomarkers to monitor tumor dynamics. Consequently, many patients undergo invasive surgical procedures to determine disease status or experience treatment delays when radiographic testing fails to show disease progression. We show here that primary CNS malignancies shed detectable levels of tumor DNA into the surrounding cerebrospinal fluid (CSF), which could serve as a sensitive and exquisitely specific marker for quantifying tumor burden without invasive biopsies. Therefore, assessment of such tumor-derived DNA in the CSF has the potential to improve the management of patients with primary CNS tumors. Cell-free DNA shed by cancer cells has been shown to be a rich source of putative tumor-specific biomarkers. Because cell-free DNA from brain and spinal cord tumors cannot usually be detected in the blood, we studied whether the cerebrospinal fluid (CSF) that bathes the CNS is enriched for tumor DNA, here termed CSF-tDNA. We analyzed 35 primary CNS malignancies and found at least one mutation in each tumor using targeted or genome-wide sequencing. Using these patient-specific mutations as biomarkers, we identified detectable levels of CSF-tDNA in 74% [95% confidence interval (95% CI) = 57–88%] of cases. All medulloblastomas, ependymomas, and high-grade gliomas that abutted a CSF space were detectable (100% of 21 cases; 95% CI = 88–100%), whereas no CSF-tDNA was detected in patients whose tumors were not directly adjacent to a CSF reservoir (P < 0.0001, Fisher’s exact test). These results suggest that CSF-tDNA could be useful for the management of patients with primary tumors of the brain or spinal cord.


Neurology | 2014

High-dose methotrexate with or without rituximab in newly diagnosed primary CNS lymphoma

Matthias Holdhoff; Prakash Ambady; Ahmed Abdelaziz; Guneet Sarai; David Bonekamp; Jaishri O. Blakeley; Stuart A. Grossman; Xiaobu Ye

Objective: To evaluate the efficacy of rituximab (R) when added to high-dose methotrexate (HD-MTX) in patients with newly diagnosed immunocompetent primary CNS lymphomas (PCNSLs). Methods: Immunocompetent adults with newly diagnosed PCNSL treated at The Johns Hopkins Hospital between 1995 and 2012 were investigated. From 1995 to 2008, patients received HD-MTX monotherapy (8 g/m2 initially every 2 weeks and after complete response [CR] monthly to complete 12 months of therapy). From 2008 to 2012, patients received the same HD-MTX with rituximab (375 mg/m2) with each HD-MTX treatment. CR rates and median overall and progression-free survival were analyzed for each patient cohort in this single-institution, retrospective study. Results: A total of 81 patients were identified: 54 received HD-MTX (median age 66 years) while 27 received HD-MTX/R (median age 65 years). CR rates were 36% in the HD-MTX cohort and 73% in the HD-MTX/R cohort (p = 0.0145). Median progression-free survival was 4.5 months in the HD-MTX cohort and 26.7 months in the HD-MTX/R cohort (p = 0.003). Median overall survival was 16.3 months in the HD-MTX cohort and has not yet been reached in the HD-MTX/R cohort (p = 0.01). Conclusions: The addition of rituximab to HD-MTX appears to improve CR rates as well as overall and progression-free survival in patients with newly diagnosed PCNSL. Comparisons of long-term survival in the 2 cohorts await further maturation of the data. Classification of evidence: This study provides Class III evidence that in immunocompetent patients with PCNSL, HD-MTX plus rituximab compared with HD-MTX alone improves CR and overall survival rates.


Journal of the National Cancer Institute | 2009

Analysis of Circulating Tumor DNA to Confirm Somatic KRAS Mutations

Matthias Holdhoff; Kerstin Schmidt; Ross C. Donehower; Luis A. Diaz

KRAS mutations have clearly emerged as a pharmacogenomic marker that can predict which metastatic colorectal cancers will be resistant to treatment with antibodies that inhibit the epidermal growth factor receptor (EGFR) ( 1 , 2 ). The evaluation of patients for mutations in KRAS is rapidly becoming part of routine practice in clinical oncology and so far has relied mostly on formalinfixed paraffin-embedded (FFPE) tumor tissue. Accurate KRAS testing is critical because it determines which patients may benefit from anti-EGFR therapy. However, the selection of specimens with a sufficient number of tumor cells, possible genetic heterogeneity between different tumor sites (eg, between primary tumor and metastases), the quality of extracted DNA, and different detection methods for KRAS mutations can interfere with accurate analysis. In addition, formalin fixation often indiscriminately and irreversibly damages DNA, increasing sample-to-sample variability and decreasing the amount of DNA available for molecular analysis. A recent article by Tol et al. ( 3 ), on the effects of KRAS mutations on first-line therapy of colorectal cancer patients with anti-EGFR therapies, highlights this issue. Eight patients had to be excluded from the study because of the discordance in the mutation status of KRAS as assessed by two independent sequencing methods, both performed on FFPE sections of tumor tissue. As an additional example, we would like to report the case of a 58-year-old man with metastatic colorectal cancer whose tumor was being evaluated for mutations in KRAS . Inadvertently, testing was performed by two independent laboratories and revealed two different results. In both laboratories, tissue sections were reviewed by a pathologist, DNA was purifi ed from the malignant areas of microdissected tumor specimens, a region of exon 2 from the KRAS [GenBank accession No. NM_004985.3] gene was amplifi ed by polymerase chain reaction (PCR) and analyzed for the presence of mutations at codons 12 and 13. The fi rst laboratory reported the presence of only wild-type KRAS by melting curve analysis. The second laboratory detected a 35G>T mutation, which causes a glycine to valine substitution at codon 12 of KRAS (G12V), using single-nucleotide primer extension analysis. To resolve these discordant fi ndings, we tested DNA from this patient’s plasma for KRAS mutations using a highly sensitive technique termed “BEAMing,” which was named after components of this method (Beads, Emulsifi cation, Amplifi cation, and Magnetics), as previously described ( 4 ). This method uses standard laboratory tools and reagents to create a water-in-oil emulsion wherein each aqueous microdroplet houses an individual fragment of DNA bound to a bead. This setting allows billions of compartmentalized PCRs to be performed in parallel in a single test tube. The products of these reactions coat each bead with thousands of copies of DNA fragments that are identical to the single DNA molecule originally present. In this case, the result is millions of beads coated entirely with either KRAS mutant or KRAS wild-type DNA. To distinguish mutant from wild-type coated beads, allele-specifi c fl uorescent probes complementary to the known wild-type or mutant sequences of KRAS are simultaneously added to the beads for hybridization. The beads are then assessed via fl ow cytometry to detect rare mutant DNA molecules among a much larger number of normal DNA molecules ( 5 ). BEAMing is a digital assay that is able to count the frequency of


Journal of The National Comprehensive Cancer Network | 2017

Central Nervous System Cancers, Version 2.2014: Featured Updates to the NCCN Guidelines

Louis B. Nabors; Jana Portnow; Mario Ammirati; Henry Brem; Paul D. Brown; Nicholas Butowski; Marc C. Chamberlain; Lisa M. DeAngelis; Robert A. Fenstermaker; Allan H. Friedman; Mark R. Gilbert; Jona A. Hattangadi-Gluth; Deneen Hesser; Matthias Holdhoff; Larry Junck; Ronald Lawson; Jay S. Loeffler; Paul L. Moots; Maciej M. Mrugala; Herbert B. Newton; Jeffrey Raizer; Lawrence Recht; Nicole Shonka; Dennis C. Shrieve; Allen K. Sills; Lode J. Swinnen; David D. Tran; Nam D. Tran; Frank D. Vrionis; Patrick Y. Wen

For many years, the diagnosis and classification of gliomas have been based on histology. Although studies including large populations of patients demonstrated the prognostic value of histologic phenotype, variability in outcomes within histologic groups limited the utility of this system. Nonetheless, histology was the only proven and widely accessible tool available at the time, thus it was used for clinical trial entry criteria, and therefore determined the recommended treatment options. Research to identify molecular changes that underlie glioma progression has led to the discovery of molecular features that have greater diagnostic and prognostic value than histology. Analyses of these molecular markers across populations from randomized clinical trials have shown that some of these markers are also predictive of response to specific types of treatment, which has prompted significant changes to the recommended treatment options for grade III (anaplastic) gliomas.


Journal of Neuro-oncology | 2013

Blood-based biomarkers for malignant gliomas

Matthias Holdhoff; Susannah G. Yovino; Osei Boadu; Stuart A. Grossman

Malignant gliomas remain incurable and present unique challenges to clinicians, radiologists and clinical and translational investigators. One of the major problems in treatment of these tumors is our limited ability to reliably assess tumor response or progression. The most frequently used neuro-imaging studies (contrast-enhanced MRI and CT) rely on changes of blood–brain barrier (BBB) integrity, providing only an indirect assessment of tumor burden. In addition, the BBB can be altered by commonly used interventions including radiation, glucocorticoids and vascular endothelial growth factor inhibitors, further complicating the interpretation of scans. Newer radiologic techniques including PET and magnetic resonance spectroscopy are theoretically promising but thus far have not meaningfully changed the assessment of patients with malignant gliomas. A tumor-specific, blood-based biomarker would be of immediate use to clinicians and investigators if sufficiently sensitive and specific. This review discusses the potential utility of such a biomarker, the general classes of tumor-derived blood-based biomarkers and it summarizes the currently available data on circulating tumor cells, circulating nucleic acids and circulating proteins in patients with malignant gliomas. It is unclear which marker or marker class appears to be the most promising for these tumors. This article provides thoughts on how novel candidate blood-based markers could be discovered and tested in a more comprehensive way and why these efforts should be among the top priorities in neuro-oncologic research in the coming years.


Journal of Neuro-oncology | 2009

Mutations of IDH1 and IDH2 are not detected in brain metastases of colorectal cancer.

Matthias Holdhoff; D. Williams Parsons; Luis A. Diaz

Somatic mutations of codon 132 of isocitrate dehydrogenase 1 (IDH1) were recently described in a genome-wide analysis of human glioblastoma multiforme (GBM) [1]. IDH1 is involved in the production of nicotinamide adenine dinucleotide phosphate (NADPH) by catalyzing the oxidative carboxylation of isocitrate to a-ketoglutarate. While only about 12% of the analyzed GBMs were found to have a mutation of IDH1, this alteration was detected in most secondary GBMs, and it appeared to be associated with comparatively younger age and improved survival. Mutations of IDH1 were also found in high frequency (over 70%) in WHO grades II and III astrocytomas and oligodendrogliomas [2, 3]. Tumors without mutations of IDH1 were often found to have mutations at the analogous position R172 of the closely related IDH2 gene [2]. These data suggest that mutations in IDH1 and IDH2 play an important role in the evolutionary, stepwise development of astrocytomas and oligodendrogliomas. Mutations of IDH, however, were not found in other solid tumors including colorectal cancer, prostate, breast and lung cancer [2, 4]. Hypothesizing that the selection pressure in the brain environment warrants certain constitutional adjustments of the cancer cells including mutational changes, we tried to address the question of whether mutations of IDH1 and IDH2 might be required for the development of brain metastasis in non-primary CNS tumors. We analyzed tissue sections of brain metastases of seven patients with metastatic colorectal cancer. The slides were reviewed together with a neuro-pathologist and areas of metastatic colorectal cancer were extracted using the lasercapture microdissection technique. The dissected material was digested overnight with proteinase K (Invitrogen), and DNA was purified using the QIAmp Micro Kit (QIAGEN). Exon 4 of the IDH1 gene and exon 4 of the IDH2 gene (containing IDH1 residue 132 and IDH2 residue 172, respectively) were PCR-amplified and sequenced as previously described [1]. None of the seven analyzed samples showed mutations in either the R132 residue of IDH1 or the R172 residue of IDH2, suggesting that these mutations are not required for the development of brain metastasis in these tumors.


The Journal of Nuclear Medicine | 2017

Repeatability of 18F-FLT PET in a Multi-Center Study of Patients with High Grade Glioma

Martin Lodge; Matthias Holdhoff; Jeffrey Leal; Asim K. Bag; L. Burt Nabors; Akiva Mintz; Glenn J. Lesser; David A. Mankoff; Arati Desai; James M. Mountz; Frank S. Lieberman; Joy D. Fisher; Serena Desideri; Xiaobu Ye; Stuart A. Grossman; David Schiff; Richard Wahl

Quantitative 3′-deoxy-3′-18F-fluorothymidine (18F-FLT) PET has potential as a noninvasive tumor biomarker for the objective assessment of response to treatment. To guide interpretation of these quantitative data, we evaluated the repeatability of 18F-FLT PET as part of a multicenter trial involving patients with high-grade glioma. Methods: 18F-FLT PET was performed on 10 patients with recurrent high-grade glioma at 5 different institutions within the Adult Brain Tumor Consortium trial ABTC1101. Data were acquired according to a double baseline protocol in which PET examinations were repeated within 2 d of each other with no intervening treatment. On each of the 2 imaging days, dedicated brain PET was performed at 2 time points, 1 and 3 h after 18F-FLT administration. Tumor SUVs and related parameters were measured at a central laboratory using various volumes of interest: isocontour at 30% of the maximum pixel (SUVmean_30%), gradient-based segmentation (SUVmean_gradient), the maximum pixel (SUVmax), and a 1-mL sphere at the region of highest uptake (SUVpeak). Repeatability coefficients (RCs) were calculated from the relative differences between corresponding SUV measurements obtained on the 2 d. Results: RCs for tumor SUVs were 22.5% (SUVmean_30%), 23.8% (SUVmean_gradient), 23.2% (SUVmax), and 18.5% (SUVpeak) at 1 h after injection. Corresponding data at 3 h were 22.4%, 25.0%, 27.3%, and 23.6%. Normalizing the tumor SUV data with reference to a background region improved repeatability, and the most stable parameter was the tumor-to-background ratio derived using SUVpeak (RC, 16.5%). Conclusion: SUV quantification of 18F-FLT uptake in glioma had an RC in the range of 18%–24% when imaging began 1 h after 18F-FLT administration. The volume-of-interest methodology had a small but not negligible influence on repeatability, with the best performance obtained using SUVpeak. Although changes in 18F-FLT SUV after treatment cannot be directly interpreted as a change in tumor proliferation, we have established ranges beyond which SUV differences are likely due to legitimate biologic effects.

Collaboration


Dive into the Matthias Holdhoff's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xiaobu Ye

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chetan Bettegowda

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Luis A. Diaz

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Bert Vogelstein

Howard Hughes Medical Institute

View shared research outputs
Top Co-Authors

Avatar

Fausto J. Rodriguez

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge