1
|
Spârchez Z, Crăciun R, Nenu I, Mocan LP, Spârchez M, Mocan T. Refining Liver Biopsy in Hepatocellular Carcinoma: An In-Depth Exploration of Shifting Diagnostic and Therapeutic Applications. Biomedicines 2023; 11:2324. [PMID: 37626820 PMCID: PMC10452389 DOI: 10.3390/biomedicines11082324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/17/2023] [Accepted: 08/19/2023] [Indexed: 08/27/2023] Open
Abstract
The field of hepatocellular carcinoma (HCC) has faced significant change on multiple levels in the past few years. The increasing emphasis on the various HCC phenotypes and the emergence of novel, specific therapies have slowly paved the way for a personalized approach to primary liver cancer. In this light, the role of percutaneous liver biopsy of focal lesions has shifted from a purely confirmatory method to a technique capable of providing an in-depth characterization of any nodule. Cancer subtype, gene expression, the mutational profile, and tissue biomarkers might soon become widely available through biopsy. However, indications, expectations, and techniques might suffer changes as the aim of the biopsy evolves from providing minimal proof of the disease to high-quality specimens for extensive analysis. Consequently, a revamped position of tissue biopsy is expected in HCC, following the reign of non-invasive imaging-only diagnosis. Moreover, given the advances in techniques that have recently reached the spotlight, such as liquid biopsy, concomitant use of all the available methods might gather just enough data to improve therapy selection and, ultimately, outcomes. The current review aims to discuss the changing role of liver biopsy and provide an evidence-based rationale for its use in the era of precision medicine in HCC.
Collapse
Affiliation(s)
- Zeno Spârchez
- Department of Gastroenterology, “Prof. Dr. O. Fodor” Regional Institute of Gastroenterology and Hepatology, 400162 Cluj-Napoca, Romania; (Z.S.); (I.N.); (T.M.)
- Department of Internal Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400162 Cluj-Napoca, Romania
| | - Rareș Crăciun
- Department of Gastroenterology, “Prof. Dr. O. Fodor” Regional Institute of Gastroenterology and Hepatology, 400162 Cluj-Napoca, Romania; (Z.S.); (I.N.); (T.M.)
- Department of Internal Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400162 Cluj-Napoca, Romania
| | - Iuliana Nenu
- Department of Gastroenterology, “Prof. Dr. O. Fodor” Regional Institute of Gastroenterology and Hepatology, 400162 Cluj-Napoca, Romania; (Z.S.); (I.N.); (T.M.)
- Department of Physiology, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
| | - Lavinia Patricia Mocan
- Department of Histology, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania;
| | - Mihaela Spârchez
- 2nd Pediatric Department, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400124 Cluj-Napoca, Romania;
| | - Tudor Mocan
- Department of Gastroenterology, “Prof. Dr. O. Fodor” Regional Institute of Gastroenterology and Hepatology, 400162 Cluj-Napoca, Romania; (Z.S.); (I.N.); (T.M.)
- UBBMed Department, Babeș-Bolyai University, 400349 Cluj-Napoca, Romania
| |
Collapse
|
2
|
Soosman SK, Schenker MP, Mazzola E, Voligny E, Smokovich A, Bay C, Nguyen T, Michael K, Jänne PA, Rabin M, Glazer DI, Johnson BE, Luo J. Safety of image guided research biopsies in patients with thoracic malignancies. Lung Cancer 2022; 173:53-57. [PMID: 36152477 DOI: 10.1016/j.lungcan.2022.08.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/09/2022] [Accepted: 08/31/2022] [Indexed: 10/31/2022]
Abstract
OBJECTIVE A common opportunity to collect research samples is during image-guided percutaneous core needle biopsies (CNBs) performed when clinically indicated or for assessing clinical trial eligibility. The relative safety of extra CNBs collected for research is undefined. MATERIALS AND METHODS Patients who underwent CNB for research purposes only [RO], as clinically indicated [CI], or as part of a clinical trial [CT] were identified. 30-day post-procedure adverse events (AEs) among the cohorts were examined and compared to the 2020 Society of Interventional Radiology QI guidelines. RESULTS 236 patients with thoracic cancers (90 % NSCLC, 5 % SCLC, 4 % mesothelioma, and 1 % thymic) had 292 CNBs (63 RO, 229 CI + CT). AEs occurred in 13 % of both the RO and CI + CT groups. Compared to the CI + CT group, the RO group did not have a higher pneumothorax incidence (RO: 5/29 [17 %], CI + CT: 18/114 [16 %], p = 0.79); both were below the suggested QI threshold of 45 % for pneumothorax. There was a negative association between number of cores obtained and risk of AE (AE vs no AE mean cores = 3.5 vs 4.8). After adjusting for the number of cores and smoking history, RO vs CI + CT lung biopsies had a higher risk of AEs (adjusted relative risk [aRR] = 2.44, 1.08-5.55, p = 0.03 vs non-lung aRR = 0.86, 0.10-7.09, p = 0.89). CONCLUSION CNBs performed for research purposes do not have a significantly increased risk of AEs when compared to those performed for clinical trials and/or when clinically indicated. However, AEs were most frequent in lung biopsies. When performing research biopsies, a target other than lung may be preferred when clinically appropriate.
Collapse
Affiliation(s)
- Steffan K Soosman
- Division of Angiography and Interventional Radiology, Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Matthew P Schenker
- Division of Angiography and Interventional Radiology, Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Emanuele Mazzola
- Lowe Center for Thoracic Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Emma Voligny
- Lowe Center for Thoracic Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Anna Smokovich
- Lowe Center for Thoracic Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Camden Bay
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Tom Nguyen
- Lowe Center for Thoracic Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kesi Michael
- Lowe Center for Thoracic Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Pasi A Jänne
- Lowe Center for Thoracic Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Michael Rabin
- Lowe Center for Thoracic Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Daniel I Glazer
- Division of Abdominal Imaging and Intervention, Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Bruce E Johnson
- Lowe Center for Thoracic Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Jia Luo
- Lowe Center for Thoracic Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
3
|
Brennan L, Brouwer-Visser J, Nüesch E, Karpova M, Heller A, Gaire F, Schneider M, Gomes B, Korski K. T-Cell Heterogeneity in Baseline Tumor Samples: Implications for Early Clinical Trial Design and Analysis. Front Immunol 2022; 13:760763. [PMID: 35558070 PMCID: PMC9086966 DOI: 10.3389/fimmu.2022.760763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 03/23/2022] [Indexed: 11/13/2022] Open
Abstract
Background In early stage clinical trials, changes to levels of tumor infiltrating lymphocytes (TILs) in the tumor microenvironment (TME) are critical biomarkers of the mechanism of action of novel immunotherapies. However, baseline heterogeneity of tumor samples, both between and within patients, and the resultant impact on the validity of clinical trial data is not well defined. Here we identify and quantify the impact of baseline variables on the heterogeneity of FoxP3+ and proliferating CD8+ T-cells levels (MKi67+CD8A+) in the TME both between and within patients for the purpose of informing clinical trial design and analysis. Methods We compared levels of FoxP3+ and MKi67+CD8+ cell densities (counts/mm2) from >1000 baseline tumor samples from clinical trials and commercially available sources. Using multivariate hierarchical regression techniques, we investigated whether inter-person heterogeneity of activated or regulatory T-cells could be attributed to baseline characteristics including demographics, indication, lesion type, tissue of excision, biopsy method, prior cancer treatment, and tissue type i.e., "fresh" or "archival" status. We also sought to characterize within-patient heterogeneity by lesion type and tissue type. Results Prior cancer treatment with hormone therapy or chemotherapy that induces immunogenic cell death may alter the TME. Archival tissue is an unreliable substitute for fresh tissue for determining baseline TIL levels. Baseline and on treatment biopsies should be matched by lesion type to avoid bias.
Collapse
Affiliation(s)
- Laura Brennan
- Roche Pharma Research and Early Development, Early Biomarker Development Oncology, Roche Innovation Center New York, Little Falls, NJ, United States
| | - Jurriaan Brouwer-Visser
- Roche Pharma Research and Early Development, Early Biomarker Development Oncology, Roche Innovation Center New York, Little Falls, NJ, United States
| | - Eveline Nüesch
- Roche Innovation Center Basel, Roche Pharma and Early Development, Basel, Switzerland
| | - Maria Karpova
- Roche Innovation Center Munich, Roche Pharma and Early Development, Penzberg, Germany
| | - Astrid Heller
- Roche Innovation Center Munich, Roche Pharma and Early Development, Penzberg, Germany
| | - Fabien Gaire
- Roche Innovation Center Munich, Roche Pharma and Early Development, Penzberg, Germany
| | - Meike Schneider
- Roche Innovation Center Basel, Roche Pharma and Early Development, Basel, Switzerland
| | - Bruno Gomes
- Roche Innovation Center Basel, Roche Pharma and Early Development, Basel, Switzerland
| | - Konstanty Korski
- Roche Innovation Center Munich, Roche Pharma and Early Development, Penzberg, Germany
| |
Collapse
|
4
|
Practical consideration for successful sequential tumor biopsies in first-in-human trials. Invest New Drugs 2022; 40:841-849. [PMID: 35404018 PMCID: PMC9288361 DOI: 10.1007/s10637-022-01236-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 03/11/2022] [Indexed: 11/23/2022]
Abstract
In first-in-human (FIH) trials, sequential tumor biopsies, i.e., two consecutive tumor biopsies, the first performed at baseline (pretreatment) and the second during the early treatment period (on-treatment), provide proof of concept in investigational new drugs. We evaluated the success of sequential tumor biopsies in FIH trials, and explored approaches for improved success rates. We retrospectively reviewed the sequential tumor biopsies required in 17 of 52 FIH trials conducted from 2015 to 2020. One hundred and thirty-eight patients were identified. Success of either pretreatment or on-treatment biopsy alone, and of sequential tumor biopsies, was defined as the acquisition of viable tumor cells and as obtaining tumor cells from both biopsy specimens, respectively. The success rates of pretreatment and on-treatment biopsy were 98.6% and 94.2%, respectively, and of sequential tumor biopsies was 70.3%. Adverse events associated with the pretreatment biopsies (33.3% positive; 72.0% negative) and timing of the first imaging assessment (before on-treatment biopsy = 40.0%; after on-treatment biopsy = 82.7%) correlated with successful sequential tumor biopsies. The reasons for unsuccessful sequential tumor biopsies could be categorized into two groups: 1) patient refusal of the on-treatment biopsy (most frequently due to early disease progression); and 2) absence of tumor cells in the pretreatment or on-treatment biopsy specimen. We propose an approach to achieving greater success in sequential tumor biopsies in FIH trials; the first imaging assessment during the study should be scheduled after on-treatment biopsy. (Registration number UMIN000042487, Date of registration November 18, 2020).
Collapse
|
5
|
Olympios N, Collet L, Paesmans M, Jungels C, Kotecki N, Awada A, Aftimos P. Analyses of the Rationale and Implementation of Research Biopsies in Oncology Clinical Trials at a Tertiary Cancer Center. Oncologist 2021; 26:1062-1070. [PMID: 34286890 PMCID: PMC8649004 DOI: 10.1002/onco.13906] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 07/09/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Biomarkers in clinical trials have led to massive incorporation of research biopsies, with potentially risks and no direct benefit for patients. In 2018, the American Society of Clinical Oncology (ASCO) released an ethical framework to provide guidance on incorporating research biopsies in cancer clinical trials. MATERIALS AND METHODS We collected biopsy requirements of cancer clinical trials conducted at Institut Jules Bordet (IJB) between 2015 and 2019 to examine adherence with the ASCO Ethical Framework. We used logistic regression models to test the association between the request for biopsy, the request for tissue, and the adherence to the ASCO framework as well as some trial characteristics. RESULTS Between January 2015 and December 2019, 178 oncological studies were conducted at IJB. Of these, 138 (78%) were sponsored by industry, 132 (74%) were phase II and III studies, and 141 (79%) concerned metastatic disease. Tissue was required for inclusion for 119 (67%) studies, among which 59 required at least one new biopsy. Adherence to ASCO's Ethical Framework was 67% for studies requiring tissue and went down to 39% for studies requiring at least one new biopsy. In multivariate analysis, requests for tissue or new biopsies increased in early-phase studies (p < .001, p < .001, respectively) and in studies investigating innovative treatments (immunotherapy or targeted therapies; p < .01, p = .02). Compliance to the ASCO framework significantly decreased with time (p < .001) and in early-phase studies (p < .001). CONCLUSION Numerous studies required tissue or new biopsies for exploratory objectives of unknown clinical utility. Requests for tissue increased over the years, whereas compliance to ASCO's Ethical Framework decreased. IMPLICATIONS FOR PRACTICE In 2019, the American Society of Clinical Oncology (ASCO) developed an ethical framework to provide guidance on incorporating research biopsies in clinical trials. This study underlines the growing request for tissue in clinical trials with potentially no impact on drug development and no benefit to actual or future patients. Adherence to ASCO's Ethical Framework decreases through time. These results highlight the importance of improving the ethics of research biopsies. ASCO's Ethical Framework offers an opportunity to improve quality of care in clinical research by maximizing scientific utility and allowing for clinically meaningful correlative science and safe access to innovative treatments for a maximum number of patients.
Collapse
Affiliation(s)
- Nathalie Olympios
- Medical Oncology Clinic, Institut Jules Bordet, Université Libre de BruxellesBrusselsBelgium
| | - Laetitia Collet
- Medical Oncology Clinic, Institut Jules Bordet, Université Libre de BruxellesBrusselsBelgium
| | - Marianne Paesmans
- Data Centre, Institut Jules Bordet, Université Libre de BruxellesBrusselsBelgium
| | - Christiane Jungels
- Medical Oncology Clinic, Institut Jules Bordet, Université Libre de BruxellesBrusselsBelgium
- Clinical Trials Conduct Unit (CTCU), Institut Jules Bordet, Université Libre de BruxellesBrusselsBelgium
| | - Nuria Kotecki
- Medical Oncology Clinic, Institut Jules Bordet, Université Libre de BruxellesBrusselsBelgium
- Clinical Trials Conduct Unit (CTCU), Institut Jules Bordet, Université Libre de BruxellesBrusselsBelgium
| | - Ahmad Awada
- Medical Oncology Clinic, Institut Jules Bordet, Université Libre de BruxellesBrusselsBelgium
| | - Philippe Aftimos
- Medical Oncology Clinic, Institut Jules Bordet, Université Libre de BruxellesBrusselsBelgium
- Clinical Trials Conduct Unit (CTCU), Institut Jules Bordet, Université Libre de BruxellesBrusselsBelgium
| |
Collapse
|
6
|
Miller L, Rosenzweig M. Biobanking: How Oncology Nurses Can Contribute to Its Use. Clin J Oncol Nurs 2021; 25:637-639. [PMID: 34800097 DOI: 10.1188/21.cjon.637-639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Biobanking is one of the most valuable tools in precision medicine. The ability of scientists to sequence tumors, blood, and normal tissue obtained from biorepositories has defined efficacious tumor targeting and a much better understanding of cancer pathology pathways. However, these biorepositories require a great deal of effort to establish and maintain. Oncology nurses are key in helping to bank tissue during routine procedures as well as complex surgeries. Nurses can obtain informed consent from patients and coordinate the banking of samples in a timely manner to ensure sample quality. Oncology nurses play an important role in informing patients of their biobanking options and connecting patients with the appropriate team for their biobanking needs.
Collapse
|
7
|
Weinfurtner K, Cho J, Ackerman D, Chen JX, Woodard A, Li W, Ostrowski D, Soulen MC, Dagli M, Shamimi-Noori S, Mondschein J, Sudheendra D, Stavropoulos SW, Reddy S, Redmond J, Khaddash T, Jhala D, Siegelman ES, Furth EE, Hunt SJ, Nadolski GJ, Kaplan DE, Gade TPF. Variability in biopsy quality informs translational research applications in hepatocellular carcinoma. Sci Rep 2021; 11:22763. [PMID: 34815453 PMCID: PMC8611010 DOI: 10.1038/s41598-021-02093-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 10/18/2021] [Indexed: 12/19/2022] Open
Abstract
In the era of precision medicine, biopsies are playing an increasingly central role in cancer research and treatment paradigms; however, patient outcomes and analyses of biopsy quality, as well as impact on downstream clinical and research applications, remain underreported. Herein, we report biopsy safety and quality outcomes for percutaneous core biopsies of hepatocellular carcinoma (HCC) performed as part of a prospective clinical trial. Patients with a clinical diagnosis of HCC were enrolled in a prospective cohort study for the genetic, proteomic, and metabolomic profiling of HCC at two academic medical centers from April 2016 to July 2020. Under image guidance, 18G core biopsies were obtained using coaxial technique at the time of locoregional therapy. The primary outcome was biopsy quality, defined as tumor fraction in the core biopsy. 56 HCC lesions from 50 patients underwent 60 biopsy events with a median of 8 core biopsies per procedure (interquartile range, IQR, 7–10). Malignancy was identified in 45/56 (80.4%, 4 without pathology) biopsy events, including HCC (40/56, 71.4%) and cholangiocarcinoma (CCA) or combined HCC-CCA (5/56, 8.9%). Biopsy quality was highly variable with a median of 40% tumor in each biopsy core (IQR 10–75). Only 43/56 (76.8%) and 23/56 (41.1%) samples met quality thresholds for genomic or metabolomic/proteomic profiling, respectively, requiring expansion of the clinical trial. Overall and major complication rates were 5/60 (8.3%) and 3/60 (5.0%), respectively. Despite uniform biopsy protocol, biopsy quality varied widely with up to 59% of samples to be inadequate for intended purpose. This finding has important consequences for clinical trial design and highlights the need for quality control prior to applications in which the presence of benign cell types may substantially alter findings.
Collapse
Affiliation(s)
- Kelley Weinfurtner
- Division of Gastroenterology and Hepatology, University of Pennsylvania, Philadelphia, PA, USA.,Penn Image-Guided Interventions Laboratory, University of Pennsylvania, Philadelphia, PA, USA
| | - Joshua Cho
- Penn Image-Guided Interventions Laboratory, University of Pennsylvania, Philadelphia, PA, USA.,Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel Ackerman
- Penn Image-Guided Interventions Laboratory, University of Pennsylvania, Philadelphia, PA, USA.,Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - James X Chen
- Vascular & Interventional Specialists of Charlotte Radiology, Charlotte, NC, USA
| | - Abashai Woodard
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Wuyan Li
- Penn Image-Guided Interventions Laboratory, University of Pennsylvania, Philadelphia, PA, USA.,Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - David Ostrowski
- Penn Image-Guided Interventions Laboratory, University of Pennsylvania, Philadelphia, PA, USA.,Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael C Soulen
- Division of Interventional Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Mandeep Dagli
- Division of Interventional Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Susan Shamimi-Noori
- Division of Interventional Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Jeffrey Mondschein
- Division of Interventional Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Deepak Sudheendra
- Division of Interventional Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Shilpa Reddy
- Division of Interventional Radiology, University of Pennsylvania, Philadelphia, PA, USA.,Corporal Michael J Cresenz VA Medical Center, Philadelphia, PA, USA
| | - Jonas Redmond
- Division of Interventional Radiology, University of Pennsylvania, Philadelphia, PA, USA.,Corporal Michael J Cresenz VA Medical Center, Philadelphia, PA, USA
| | - Tamim Khaddash
- Division of Interventional Radiology, University of Pennsylvania, Philadelphia, PA, USA.,Corporal Michael J Cresenz VA Medical Center, Philadelphia, PA, USA
| | - Darshana Jhala
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Evan S Siegelman
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Emma E Furth
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephen J Hunt
- Penn Image-Guided Interventions Laboratory, University of Pennsylvania, Philadelphia, PA, USA.,Division of Interventional Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Gregory J Nadolski
- Penn Image-Guided Interventions Laboratory, University of Pennsylvania, Philadelphia, PA, USA.,Corporal Michael J Cresenz VA Medical Center, Philadelphia, PA, USA
| | - David E Kaplan
- Division of Gastroenterology and Hepatology, University of Pennsylvania, Philadelphia, PA, USA.,Penn Image-Guided Interventions Laboratory, University of Pennsylvania, Philadelphia, PA, USA.,Corporal Michael J Cresenz VA Medical Center, Philadelphia, PA, USA
| | - Terence P F Gade
- Penn Image-Guided Interventions Laboratory, University of Pennsylvania, Philadelphia, PA, USA. .,Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA. .,Division of Interventional Radiology, University of Pennsylvania, Philadelphia, PA, USA. .,Corporal Michael J Cresenz VA Medical Center, Philadelphia, PA, USA. .,Radiology and Cancer Biology, University of Pennsylvania Perelman School of Medicine, 652 BRB II/III, 421 Curie Blvd, Philadelphia, PA, 19104-6160, USA.
| |
Collapse
|
8
|
Enhanced Detection of Desmoplasia by Targeted Delivery of Iron Oxide Nanoparticles to the Tumour-Specific Extracellular Matrix. Pharmaceutics 2021; 13:pharmaceutics13101663. [PMID: 34683956 PMCID: PMC8539756 DOI: 10.3390/pharmaceutics13101663] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 09/27/2021] [Accepted: 09/30/2021] [Indexed: 01/14/2023] Open
Abstract
Diagnostic imaging of aggressive cancer with a high stroma content may benefit from the use of imaging contrast agents targeted with peptides that have high binding affinity to the extracellular matrix (ECM). In this study, we report the use of superparamagnetic iron-oxide nanoparticles (IO-NP) conjugated to a nonapeptide, CSGRRSSKC (CSG), which specifically binds to the laminin-nidogen-1 complex in tumours. We show that CSG-IO-NP accumulate in tumours, predominantly in the tumour ECM, following intravenous injection into a murine model of pancreatic neuroendocrine tumour (PNET). In contrast, a control untargeted IO-NP consistently show poor tumour uptake, and IO-NP conjugated to a pentapeptide, CREKA that bind fibrin clots in blood vessels show restricted uptake in the angiogenic vessels of the tumours. CSG-IO-NP show three-fold higher intratumoral accumulation compared to CREKA-IO-NP. Magnetic resonance imaging (MRI) T2-weighted scans and T2 relaxation times indicate significant uptake of CSG-IO-NP irrespective of tumour size, whereas the uptake of CREKA-IO-NP is only consistent in small tumours of less than 3 mm in diameter. Larger tumours with significantly reduced tumour blood vessels show a lack of CREKA-IO-NP uptake. Our data suggest CSG-IO-NP are particularly useful for detecting stroma in early and advanced solid tumours.
Collapse
|
9
|
Bruinsma RS, Nievelstein RAJ, Littooij AS, Vermeulen MA, van de Ven CP, van Noesel MM, Wijnen MHWA, van der Steeg AFW, de Krijger RR. Diagnostic accuracy of image-guided core needle biopsy of non-central nervous system tumors in children. Pediatr Blood Cancer 2021; 68:e29179. [PMID: 34121329 DOI: 10.1002/pbc.29179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 11/08/2022]
Abstract
BACKGROUND AND AIMS Core needle biopsies (CNB) are less invasive, cause less morbidity, and have lower costs than open biopsies (OB). However, the number of studies reporting CNB accuracy in pediatric tumors is limited and series are small. The aim of this study is to investigate if CNB diagnosis is concordant with the final diagnosis in pediatric solid non-central nervous system (CNS) tumors. METHODS Data from all patients treated in a single center between November 2014 and December 2019 were collected from the national pathology database and from local medical records. Data collection included age, sex, CNB diagnosis, final diagnosis, number of cores obtained, number of cores used for histology, cumulative core length, greatest dimension of the lesion, lesion volume, and complications. RESULTS Out of 361 CNB, 95.6% (345/361) provided a diagnosis. A resection or follow-up biopsy was performed in 201 cases. The final diagnosis was concordant with the CNB in 100% (201/201) of cases. The age, number of cores used for histology, and the greatest dimension of the lesion did not significantly differ between diagnostic and nondiagnostic CNB. The cumulative core length of diagnostic CNB was significantly higher than in the nondiagnostic group (24.72 mm vs. 13.37 mm, p-value .022). Complications occurred in 2.1% (7/337) of CNB procedures. Molecular analysis was successful in 228/233 (98%) of cases in which it was performed. CONCLUSIONS CNB diagnosis is highly concordant with the final diagnosis and the diagnostic rate is high. The complication rate in CNB is low.
Collapse
Affiliation(s)
- Rixt S Bruinsma
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Rutger A J Nievelstein
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.,Department of Radiology & Nuclear Medicine, Division Imaging & Oncology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Annemieke S Littooij
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.,Department of Radiology & Nuclear Medicine, Division Imaging & Oncology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | | | | | - Max M van Noesel
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Marc H W A Wijnen
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | | | - Ronald R de Krijger
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.,Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
10
|
Gastman B, Agarwal PK, Berger A, Boland G, Broderick S, Butterfield LH, Byrd D, Fecci PE, Ferris RL, Fong Y, Goff SL, Grabowski MM, Ito F, Lim M, Lotze MT, Mahdi H, Malafa M, Morris CD, Murthy P, Neves RI, Odunsi A, Pai SI, Prabhakaran S, Rosenberg SA, Saoud R, Sethuraman J, Skitzki J, Slingluff CL, Sondak VK, Sunwoo JB, Turcotte S, Yeung CC, Kaufman HL. Defining best practices for tissue procurement in immuno-oncology clinical trials: consensus statement from the Society for Immunotherapy of Cancer Surgery Committee. J Immunother Cancer 2020; 8:e001583. [PMID: 33199512 PMCID: PMC7670953 DOI: 10.1136/jitc-2020-001583] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2020] [Indexed: 12/11/2022] Open
Abstract
Immunotherapy is now a cornerstone for cancer treatment, and much attention has been placed on the identification of prognostic and predictive biomarkers. The success of biomarker development is dependent on accurate and timely collection of biospecimens and high-quality processing, storage and shipping. Tumors are also increasingly used as source material for the generation of therapeutic T cells. There have been few guidelines or consensus statements on how to optimally collect and manage biospecimens and source material being used for immunotherapy and related research. The Society for Immunotherapy of Cancer Surgery Committee has brought together surgical experts from multiple subspecialty disciplines to identify best practices and to provide consensus on how best to access and manage specific tissues for immuno-oncology treatments and clinical investigation. In addition, the committee recommends early integration of surgeons and other interventional physicians with expertise in biospecimen collection, especially in clinical trials, to optimize the quality of tissue and the validity of correlative clinical studies in cancer immunotherapy.
Collapse
Affiliation(s)
- Brian Gastman
- Department of Plastic Surgery, Cleveland Clinic, Cleveland, Ohio, USA
| | - Piyush K Agarwal
- Department of Surgery, University of Chicago, Chicago, Illinois, USA
| | - Adam Berger
- Division of Surgical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
| | - Genevieve Boland
- Department of Surgical Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Stephen Broderick
- Oncology, Johns Hopkins Medicine Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland, USA
- Department of Surgery, Johns Hopkins Medicine, Baltimore, Maryland, USA
| | - Lisa H Butterfield
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
- Microbiology and Immunology, University of California San Francisco, San Francisco, California, USA
| | - David Byrd
- Department of Surgery, University of Washington, Seattle, Washington, USA
| | - Peter E Fecci
- Department of Neurosurgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Robert L Ferris
- Departments of Otolaryngology, Immunology, and Radiation Oncology, University of Pittsburgh Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Yuman Fong
- Department of Surgery, City of Hope National Medical Center, Duarte, California, USA
| | | | - Matthew M Grabowski
- Department of Neurosurgery, Duke Center for Brain and Spine Metastasis, Durham, North Carolina, USA
| | - Fumito Ito
- Center for Immunotherapy, Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Michael Lim
- Departments of Neurosurgery, Oncology, Radiation Oncology, and Otolaryngology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Michael T Lotze
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Haider Mahdi
- OBGYN and Women's Health Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Mokenge Malafa
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Carol D Morris
- Division of Orthopaedic Oncology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Pranav Murthy
- Department of Surgery, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Rogerio I Neves
- Department of Surgery, Penn State Cancer Institute, Hershey, Pennsylvania, USA
| | - Adekunle Odunsi
- Departments of Immunology and Gynecologic Oncology, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Sara I Pai
- Department of Surgical Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Sangeetha Prabhakaran
- Division of Surgical Oncology, Department of Surgery, UNM Comprehensive Cancer Center, University of New Mexico, Albuquerque, New Mexico, USA
| | | | - Ragheed Saoud
- Department of Surgery, University of Chicago Hospitals, Chicago, Illinois, United States
| | | | - Joseph Skitzki
- Departments of Surgical Oncology and Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Craig L Slingluff
- Department of Surgery, Division of Surgical Oncology, Breast and Melanoma Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Vernon K Sondak
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| | - John B Sunwoo
- Department of Otolaryngology, Stanford University School of Medicine, Stanford, California, USA
| | - Simon Turcotte
- Surgery Department, Centre Hospitalier de l'Universite de Montreal, Montreal, Quebec, Canada
| | - Cecilia Cs Yeung
- Department of Pathology, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Howard L Kaufman
- Department of Surgical Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Immuneering Corp, Cambridge, Massachusetts, USA
| |
Collapse
|