51
|
Dall'Olio FG, Marabelle A, Caramella C, Garcia C, Aldea M, Chaput N, Robert C, Besse B. Tumour burden and efficacy of immune-checkpoint inhibitors. Nat Rev Clin Oncol 2021; 19:75-90. [PMID: 34642484 DOI: 10.1038/s41571-021-00564-3] [Citation(s) in RCA: 178] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2021] [Indexed: 01/07/2023]
Abstract
Accumulating evidence suggests that a high tumour burden has a negative effect on anticancer immunity. The concept of tumour burden, simply defined as the total amount of cancer in the body, in contrast to molecular tumour burden, is often poorly understood by the wider medical community; nonetheless, a possible role exists in defining the optimal treatment strategy for many patients. Historically, tumour burden has been assessed using imaging. In particular, CT scans have been used to evaluate both the number and size of metastases as well as the number of organs involved. These methods are now often complemented by metabolic tumour burden, measured using the more recently developed 2-deoxy-2-[18F]-fluoro-D-glucose (FDG)-PET/CT. Serum-based biomarkers, such as lactate dehydrogenase, can also reflect tumour burden and are often also correlated with a poor response to immune-checkpoint inhibitors. Other circulating markers (such as circulating free tumour DNA and/or circulating tumour cells) are also attracting research interest as surrogate markers of tumour burden. In this Review, we summarize evidence supporting the utility of tumour burden as a biomarker to guide the use of immune-checkpoint inhibitors. We also describe data and provide perspective on the various tools used for tumour burden assessment, with a particular emphasis on future therapeutic strategies that might address the issue of inferior outcomes among patients with cancer with a high tumour burden.
Collapse
Affiliation(s)
- Filippo G Dall'Olio
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France.,Division of Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Specialized, Experimental and Diagnostic Medicine, University of Bologna, Bologna, Italy
| | - Aurélien Marabelle
- Drug Development Department, Gustave Roussy, Villejuif, France.,Faculty of Medicine, University Paris-Saclay, Kremlin Bicêtre, France.,Institut national de la santé et de la recherche médicale (INSERM), Gustave Roussy, Villejuif, France
| | - Caroline Caramella
- Department of Radiology, Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - Camilo Garcia
- Department of Nuclear Medicine and Endocrine Oncology, Institut Gustave Roussy and University Paris-Saclay, Villejuif, France
| | - Mihaela Aldea
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| | - Nathalie Chaput
- Laboratory of Immunomonitoring in Oncology, Gustave Roussy, Villejuif, France.,Faculty of Pharmacy, University Paris-Saclay, Chatenay-Malabry, France
| | - Caroline Robert
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France.,Faculty of Medicine, University Paris-Saclay, Kremlin Bicêtre, France.,Institut national de la santé et de la recherche médicale (INSERM), Gustave Roussy, Villejuif, France
| | - Benjamin Besse
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France. .,Faculty of Medicine, University Paris-Saclay, Kremlin Bicêtre, France.
| |
Collapse
|
52
|
Abstract
Biological allometries, such as the scaling of metabolism to mass, are hypothesized to result from natural selection to maximize how vascular networks fill space yet minimize internal transport distances and resistance to blood flow. Metabolic scaling theory argues two guiding principles—conservation of fluid flow and space-filling fractal distributions—describe a diversity of biological networks and predict how the geometry of these networks influences organismal metabolism. Yet, mostly absent from past efforts are studies that directly, and independently, measure metabolic rate from respiration and vascular architecture for the same organ, organism, or tissue. Lack of these measures may lead to inconsistent results and conclusions about metabolism, growth, and allometric scaling. We present simultaneous and consistent measurements of metabolic scaling exponents from clinical images of lung cancer, serving as a first-of-its-kind test of metabolic scaling theory, and identifying potential quantitative imaging biomarkers indicative of tumor growth. We analyze data for 535 clinical PET-CT scans of patients with non-small cell lung carcinoma to establish the presence of metabolic scaling between tumor metabolism and tumor volume. Furthermore, we use computer vision and mathematical modeling to examine predictions of metabolic scaling based on the branching geometry of the tumor-supplying blood vessel networks in a subset of 56 patients diagnosed with stage II-IV lung cancer. Examination of the scaling of maximum standard uptake value with metabolic tumor volume, and metabolic tumor volume with gross tumor volume, yield metabolic scaling exponents of 0.64 (0.20) and 0.70 (0.17), respectively. We compare these to the value of 0.85 (0.06) derived from the geometric scaling of the tumor-supplying vasculature. These results: (1) inform energetic models of growth and development for tumor forecasting; (2) identify imaging biomarkers in vascular geometry related to blood volume and flow; and (3) highlight unique opportunities to develop and test the metabolic scaling theory of ecology in tumors transitioning from avascular to vascular geometries.
Collapse
|
53
|
Nobashi TW, Mayer AT, Xiao Z, Chan CT, Chaney AM, James ML, Gambhir SS. Whole-body PET Imaging of T-cell Response to Glioblastoma. Clin Cancer Res 2021; 27:6445-6456. [PMID: 34548318 DOI: 10.1158/1078-0432.ccr-21-1412] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/30/2021] [Accepted: 09/17/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Immunotherapy is a promising approach for many oncological malignancies, including glioblastoma, however, there are currently no available tools or biomarkers to accurately assess whole-body immune responses in patients with glioblastoma treated with immunotherapy. Here, the utility of OX40, a costimulatory molecule mainly expressed on activated effector T cells known to play an important role in eliminating cancer cells, was evaluated as a PET imaging biomarker to quantify and track response to immunotherapy. EXPERIMENTAL DESIGN A subcutaneous vaccination approach of CpG oligodeoxynucleotide, OX40 mAb, and tumor lysate at a remote site in a murine orthotopic glioma model was developed to induce activation of T cells distantly while monitoring their distribution in stimulated lymphoid organs with respect to observed therapeutic effects. To detect OX40-positive T cells, we utilized our in-house-developed 89Zr-DFO-OX40 mAb and in vivo PET/CT imaging. RESULTS ImmunoPET with 89Zr-DFO-OX40 mAb revealed strong OX40-positive responses with high specificity, not only in the nearest lymph node from vaccinated area (mean, 20.8%ID/cc) but also in the spleen (16.7%ID/cc) and the tumor draining lymph node (11.4%ID/cc). When the tumor was small (<106 p/sec/cm2/sr in bioluminescence imaging), a high number of responders and percentage shrinkage in tumor signal was indicated after only a single cycle of vaccination. CONCLUSIONS The results highlight the promise of clinically translating cancer vaccination as a potential glioma therapy, as well as the benefits of monitoring efficacy of these treatments using immunoPET imaging of T-cell activation.
Collapse
Affiliation(s)
- Tomomi W Nobashi
- Department of Radiology, Stanford University, Stanford, California.
| | - Aaron T Mayer
- Department of Radiology, Stanford University, Stanford, California. .,Department of Bioengineering, Stanford University, Stanford, California.,Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California.,Bio-X Program at Stanford, Stanford University, Stanford, California
| | - Zunyu Xiao
- Department of Radiology, Stanford University, Stanford, California.,Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California.,Molecular Imaging Research Center of Harbin Medical University, Harbin, China
| | - Carmel T Chan
- Department of Radiology, Stanford University, Stanford, California.,Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California
| | - Aisling M Chaney
- Department of Radiology, Stanford University, Stanford, California.,Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California
| | - Michelle L James
- Department of Radiology, Stanford University, Stanford, California.,Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California
| | - Sanjiv S Gambhir
- Department of Radiology, Stanford University, Stanford, California.,Department of Bioengineering, Stanford University, Stanford, California.,Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California.,Bio-X Program at Stanford, Stanford University, Stanford, California.,Department of Materials Science and Engineering, Stanford University, Stanford, California.,Canary Center at Stanford, Stanford University, Stanford, California
| |
Collapse
|
54
|
Immune Checkpoint Inhibitors in Advanced NSCLC: [ 18F]FDG PET/CT as a Troubleshooter in Treatment Response. Diagnostics (Basel) 2021; 11:diagnostics11091681. [PMID: 34574022 PMCID: PMC8471751 DOI: 10.3390/diagnostics11091681] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/02/2021] [Accepted: 09/11/2021] [Indexed: 12/17/2022] Open
Abstract
Introduction: The aim of this study was to investigate whether [18F]FDG PET/CT-derived semi-quantitative parameters can predict immunotherapy treatment response in non-small cell lung cancer (NSCLC) patients. Secondly, immune-related adverse events (irAEs) and lymphoid cell-rich organs activation were evaluated. Materials and Methods: Twenty-eight patients who underwent [18F]FDG PET/CT scans before and at first restaging therapy with immuno-checkpoint inhibitors (ICIs) were retrospectively analyzed. PET-based semi-quantitative parameters extracted from both scans were respectively: SUVmax and SUVpeak of the target lesion, whole-body metabolic tumor volume (MTVWB), and whole-body total lesion glycolysis (TLGWB), as well as their interval changes (ΔSUVmaxTL, ΔSUVpeakTL, ΔMTVWB, ΔTLGWB). These PET-derived parameters were correlated to controlled disease (CD) assessed by RECIST 1.1. IrAEs, if present, were also described and correlated with clinical benefit (CB). SUVmax of the spleen and bone marrow at restaging scans were also correlated to CB. Results: The CD was achieved in 54% of patients. Out of 28 eligible patients, 13 (46%) experienced progressive disease (PD), 7 showed SD, 7 had PR, and only in one patient CR was achieved. ΔSUVmaxTL (p = 0.002) and ΔSUVpeakTL (p < 0.001) as well as ΔMTVWB (p < 0.001) and ΔTLGWB (p < 0.005) were significantly associated with PD vs. non-PD. IrAEs and lymphoid cell-rich organs activation did not correlate with CB. Conclusions: [18F]FDG PET/CT by using interval changes of PET-derived semi-quantitative parameters could represent a reliable tool in immunotherapy treatment response evaluation in NSCLC patients.
Collapse
|
55
|
Leung D, Bonacorsi S, Smith RA, Weber W, Hayes W. Molecular Imaging and the PD-L1 Pathway: From Bench to Clinic. Front Oncol 2021; 11:698425. [PMID: 34497758 PMCID: PMC8420047 DOI: 10.3389/fonc.2021.698425] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 07/22/2021] [Indexed: 01/24/2023] Open
Abstract
Programmed death-1 (PD-1) and programmed death ligand 1 (PD-L1) inhibitors target the important molecular interplay between PD-1 and PD-L1, a key pathway contributing to immune evasion in the tumor microenvironment (TME). Long-term clinical benefit has been observed in patients receiving PD-(L)1 inhibitors, alone and in combination with other treatments, across multiple tumor types. PD-L1 expression has been associated with response to immune checkpoint inhibitors, and treatment strategies are often guided by immunohistochemistry-based diagnostic tests assessing expression of PD-L1. However, challenges related to the implementation, interpretation, and clinical utility of PD-L1 diagnostic tests have led to an increasing number of preclinical and clinical studies exploring interrogation of the TME by real-time imaging of PD-(L)1 expression by positron emission tomography (PET). PET imaging utilizes radiolabeled molecules to non-invasively assess PD-(L)1 expression spatially and temporally. Several PD-(L)1 PET tracers have been tested in preclinical and clinical studies, with clinical trials in progress to assess their use in a number of cancer types. This review will showcase the development of PD-(L)1 PET tracers from preclinical studies through to clinical use, and will explore the opportunities in drug development and possible future clinical implementation.
Collapse
Affiliation(s)
- David Leung
- Translational Medicine, Bristol Myers Squibb, Princeton, NJ, United States
| | - Samuel Bonacorsi
- Translational Medicine, Bristol Myers Squibb, Princeton, NJ, United States
| | - Ralph Adam Smith
- Translational Medicine, Bristol Myers Squibb, Princeton, NJ, United States
| | - Wolfgang Weber
- Technische Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Wendy Hayes
- Translational Medicine, Bristol Myers Squibb, Princeton, NJ, United States
| |
Collapse
|
56
|
Lenci E, Marcantognini G, Cognigni V, Lupi A, Rinaldi S, Cantini L, Fiordoliva I, Carloni AL, Rocchi M, Zuccatosta L, Gasparini S, Berardi R. Tumor burden as possible biomarker of outcome in advanced NSCLC patients treated with immunotherapy: a single center, retrospective, real-world analysis. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2021; 2:227-239. [PMID: 36046436 PMCID: PMC9400786 DOI: 10.37349/etat.2021.00043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 04/19/2021] [Indexed: 11/19/2022] Open
Abstract
AIM The role of tumor burden (TB) for patients with non-small cell lung cancer (NSCLC) receiving immunotherapy is still unknown. The aim of this analysis was to analyze the prognostic value of TB in a real-world sample of advanced NSCLC patients. METHODS Sixty-five consecutive patients with advanced NSCLC treated with immunotherapy as first or second line therapy were retrospectively analyzed between August 2015 and February 2018. TB was recorded at baseline considering sites and number of metastases, thoracic vs. extrathoracic disease, measurable disease (MD) vs. not-MD (NMD) and evaluating dimensional aspects as maximum lesion diameter (cut-off = 6.3 cm), sum of the 5 major lesions diameters (cut-off = 14.3 cm), and number of sites of metastases (cut-off > 4). All cut-offs were calculated by receiver operating characteristic curves. Median overall survival (OS) was estimated using Kaplan-Meier method. A Cox regression model was carried out for univariate and multivariate analyses. RESULTS Median age was 70 years and most patients (86.2%) had a good performance status (PS-Eastern Cooperative Oncology Group < 2). No significant difference in OS was noted between subgroups of patients according to TB. Bone metastases (BM) had a negative prognostic impact [median OS (mOS), 13.8 vs. 70.0 months, P = 0.0009; median progression free survival in the second line (mPFS2) 2.97 vs. 8.63 months; P = 0.0037]. Patients with NMD had a poorer prognosis (mOS, 15.9 months vs. not reached, P < 0.0001; mPFS2 3.8 vs. 12.2 months; P = 0.0199). Patients with disease limited to the thorax had a better prognosis compared to patients with involvement of extrathoracic sites (mOS, 70 vs. 17.3 months; P = 0.0136). Having more than 4 metastatic sites resulted as a negative prognostic factor (mOS, 15.9 vs. 25.2 months; P = 0.0106). At multivariate analysis, BM, NMD, extrathoracic disease and number of sites of metastases > 4 were negative prognostic factors (P < 0.0001). CONCLUSIONS This study underlines the negative prognostic impact of specific metastatic sites, presence of NMD and extrathoracic disease in advanced NSCLC patients treated with immunotherapy. However, TB does not appear to affect the outcome of these patients.
Collapse
Affiliation(s)
- Edoardo Lenci
- Clinical Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti, 60126 Ancona, Italy
| | - Giulia Marcantognini
- Clinical Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti, 60126 Ancona, Italy
| | - Valeria Cognigni
- Clinical Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti, 60126 Ancona, Italy
| | - Alessio Lupi
- Clinical Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti, 60126 Ancona, Italy
| | - Silvia Rinaldi
- Clinical Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti, 60126 Ancona, Italy
| | - Luca Cantini
- Clinical Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti, 60126 Ancona, Italy
| | - Ilaria Fiordoliva
- Clinical Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti, 60126 Ancona, Italy
| | - Anna Lisa Carloni
- Clinical Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti, 60126 Ancona, Italy
| | - Marco Rocchi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Lina Zuccatosta
- Operative Unit of Pneumology, Ospedali Riuniti University Hospital, 60126 Ancona, Italy
| | - Stefano Gasparini
- Operative Unit of Pneumology, Ospedali Riuniti University Hospital, 60126 Ancona, Italy
| | - Rossana Berardi
- Clinical Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti, 60126 Ancona, Italy
| |
Collapse
|
57
|
Pieper AA, Rakhmilevich AL, Spiegelman DV, Patel RB, Birstler J, Jin WJ, Carlson PM, Charych DH, Hank JA, Erbe AK, Overwijk WW, Morris ZS, Sondel PM. Combination of radiation therapy, bempegaldesleukin, and checkpoint blockade eradicates advanced solid tumors and metastases in mice. J Immunother Cancer 2021; 9:jitc-2021-002715. [PMID: 34172518 PMCID: PMC8237721 DOI: 10.1136/jitc-2021-002715] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2021] [Indexed: 01/11/2023] Open
Abstract
Background Current clinical trials are using radiation therapy (RT) to enhance an antitumor response elicited by high-dose interleukin (IL)-2 therapy or immune checkpoint blockade (ICB). Bempegaldesleukin (BEMPEG) is an investigational CD122-preferential IL-2 pathway agonist with prolonged in vivo half-life and preferential intratumoral expansion of T effector cells over T regulatory cells. BEMPEG has shown encouraging safety and efficacy in clinical trials when used in combination with PD-1 checkpoint blockade. In this study, we investigated the antitumor effect of local RT combined with BEMPEG in multiple immunologically ‘cold’ tumor models. Additionally, we asked if ICB could further enhance the local and distant antitumor effect of RT+BEMPEG in the setting of advanced solid tumors or metastatic disease. Methods Mice bearing flank tumors (B78 melanoma, 4T1 breast cancer, or MOC2 head and neck squamous cell carcinoma) were treated with combinations of RT and immunotherapy (including BEMPEG, high-dose IL-2, anti(α)-CTLA-4, and α-PD-L1). Mice bearing B78 flank tumors were injected intravenously with B16 melanoma cells to mimic metastatic disease and were subsequently treated with RT and/or immunotherapy. Tumor growth and survival were monitored. Peripheral T cells and tumor-infiltrating lymphocytes were assessed via flow cytometry. Results A cooperative antitumor effect was observed in all models when RT was combined with BEMPEG, and RT increased IL-2 receptor expression on peripheral T cells. This cooperative interaction was associated with increased IL-2 receptor expression on peripheral T cells following RT. In the B78 melanoma model, RT+BEMPEG resulted in complete tumor regression in the majority of mice with a single ~400 mm3 tumor. This antitumor response was T-cell dependent and supported by long-lasting immune memory. Adding ICB to RT+BEMPEG strengthened the antitumor response and cured the majority of mice with a single ~1000 mm3 B78 tumor. In models with disseminated metastasis (B78 primary with B16 metastasis, 4T1, and MOC2), the triple combination of RT, BEMPEG, and ICB significantly improved primary tumor response and survival. Conclusion The combination of local RT, BEMPEG, and ICB cured mice with advanced, immunologically cold tumors and distant metastasis in a T cell-dependent manner, suggesting this triple combination warrants clinical testing.
Collapse
Affiliation(s)
- Alexander A Pieper
- Department of Human Oncology, University of Wisconsin Madison, Madison, Wisconsin, USA
| | | | - Daniel V Spiegelman
- Department of Human Oncology, University of Wisconsin Madison, Madison, Wisconsin, USA
| | - Ravi B Patel
- Department of Radiation Oncology, University of Pittsburgh Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Jen Birstler
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Won Jong Jin
- Department of Human Oncology, University of Wisconsin Madison, Madison, Wisconsin, USA
| | - Peter M Carlson
- Department of Human Oncology, University of Wisconsin Madison, Madison, Wisconsin, USA
| | | | - Jacquelyn A Hank
- Department of Human Oncology, University of Wisconsin Madison, Madison, Wisconsin, USA
| | - Amy K Erbe
- Department of Human Oncology, University of Wisconsin Madison, Madison, Wisconsin, USA
| | | | - Zachary S Morris
- Department of Human Oncology, University of Wisconsin Madison, Madison, Wisconsin, USA
| | - Paul M Sondel
- Department of Human Oncology, University of Wisconsin Madison, Madison, Wisconsin, USA .,Department of Pediatrics, University of Wisconsin Madison, Madison, Wisconsin, USA
| |
Collapse
|
58
|
Dumoulin DW, Cornelissen R, Bezemer K, Baart SJ, Aerts JGJV. Long-Term Follow-Up of Mesothelioma Patients Treated with Dendritic Cell Therapy in Three Phase I/II Trials. Vaccines (Basel) 2021; 9:vaccines9050525. [PMID: 34069348 PMCID: PMC8158710 DOI: 10.3390/vaccines9050525] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/14/2021] [Accepted: 05/17/2021] [Indexed: 12/29/2022] Open
Abstract
Background: Malignant pleural mesothelioma (MPM) is a fatal neoplasm with, if untreated, poor survival of approximately nine months from diagnosis. Until recently, phase II–III immunotherapy trials did not show any significant benefit. The lack of immunotherapy efficacy can be explained by the fact that mesothelioma is a tumor with an “immune desert” phenotype, meaning a non-inflamed tumor characterized by low T-cell infiltration. By administration of DCs, which were ex-vivo cultured, exposed to (tumor-associated) antigens, and subsequently activated, this “immune desert” phenotype might be turned into an “inflamed” phenotype. Three phase I/II studies have been performed and published using activated DCs, which support this concept. We here report on the long-term survival of patients treated with DCs in three phase I/II studies. Methods: Survival data of the phase I/II trials using DC therapy in MPM patients were obtained and subsequently analyzed. In the first two trials, DCs were loaded with autologous tumor lysate. In the third trial, DCs were loaded with allogeneic mesothelioma tumor cell line lysate. Results: In the three studies combined, 29 patients with MPM were treated with DC vaccination between 2006 and 2015. At data cut-off, the median OS was 27 months (95% CI: 21–47 months). OS at 2 years was 55.2% (95% CI: 39.7–76.6%), and OS at 5 years was 20.7% (95% CI: 10.1–42.2%). Conclusions: The long-term survival of DC therapy in MPM in these three trials is promising, which is the basis for the randomized phase II/III DENIM study. This DENIM study is currently enrolling, and the results of which have to be awaited for definite conclusions.
Collapse
Affiliation(s)
- Daphne W. Dumoulin
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands; (R.C.); (K.B.); (J.G.J.V.A.)
- Correspondence:
| | - Robin Cornelissen
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands; (R.C.); (K.B.); (J.G.J.V.A.)
| | - Koen Bezemer
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands; (R.C.); (K.B.); (J.G.J.V.A.)
| | - Sara J. Baart
- Department of Biostatistics, Erasmus MC, 3015 GD Rotterdam, The Netherlands;
| | - Joachim G. J. V. Aerts
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands; (R.C.); (K.B.); (J.G.J.V.A.)
| |
Collapse
|
59
|
Monaco L, Gemelli M, Gotuzzo I, Bauckneht M, Crivellaro C, Genova C, Cortinovis D, Zullo L, Ammoni LC, Bernasconi DP, Rossi G, Morbelli S, Guerra L. Metabolic Parameters as Biomarkers of Response to Immunotherapy and Prognosis in Non-Small Cell Lung Cancer (NSCLC): A Real World Experience. Cancers (Basel) 2021; 13:1634. [PMID: 33915801 PMCID: PMC8037395 DOI: 10.3390/cancers13071634] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/26/2021] [Accepted: 03/29/2021] [Indexed: 12/22/2022] Open
Abstract
Immune-checkpoint inhibitors (ICIs) have been proven to have great efficacy in non-small cell lung cancer (NSCLC) as single agents or in combination therapy, being capable to induce deep and durable remission. However, severe adverse events may occur and about 40% of patients do not benefit from the treatment. Predictive factors of response to ICIs are needed in order to customize treatment. The aim of this study is to evaluate the correlation between quantitative positron emission tomography (PET) parameters defined before starting ICI therapy and responses to treatment and patient outcome. We retrospectively analyzed 92 NSCLC patients treated with nivolumab, pembrolizumab or atezolizumab. Basal PET/computed tomography (CT) scan parameters (whole-body metabolic tumor volume-wMTV, total lesion glycolysis-wTLG, higher standardized uptake volume maximum and mean-SUVmax and SUVmean) were calculated for each patient and correlated with outcomes. Patients who achieved disease control (complete response + partial response + stable disease) had significantly lower MTV median values than patients who had not (progressive disease) (77 vs. 160.2, p = 0.039). Furthermore, patients with MTV and TLG values lower than the median values had improved OS compared to patients with higher MTV and TLG (p = 0.03 and 0.05, respectively). No relation was found between the other parameters and outcome. In conclusion, baseline metabolic tumor burden, measured with MTV, might be an independent predictor of treatment response to ICI and a prognostic biomarker in NSCLC patients.
Collapse
Affiliation(s)
- Lavinia Monaco
- School of Medicine and Surgery, University of Milano Bicocca, 20900 Monza, Italy; (L.M.); (L.G.)
| | - Maria Gemelli
- Medical Oncology, ASST Monza, San Gerardo Hospital, 20900 Monza, Italy; (M.G.); (D.C.)
| | - Irene Gotuzzo
- School of Medicine and Surgery, University of Milano Bicocca, 20900 Monza, Italy; (L.M.); (L.G.)
| | - Matteo Bauckneht
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.B.); (S.M.)
- Nuclear Medicine Unit, Department of Health Sciences, University of Genoa, 16132 Genoa, Italy
| | - Cinzia Crivellaro
- Nuclear Medicine, ASST Monza San Gerardo Hospital, 20900 Monza, Italy;
| | - Carlo Genova
- UOC Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy;
- Dipartimento di Medicina Interna e Specialità Mediche (DiMI), Facoltà di Medicina e Chirurgia, Università degli Studi di Genova, 16132 Genova, Italy
| | - Diego Cortinovis
- Medical Oncology, ASST Monza, San Gerardo Hospital, 20900 Monza, Italy; (M.G.); (D.C.)
| | - Lodovica Zullo
- UOC Oncologia Medica 2, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy;
| | | | - Davide Paolo Bernasconi
- Bicocca Biostatistics Bioinformatics and Bioimaging Center—B4, School of Medicine and Surgery, University Milano Bicocca, 20128 Milano, Italy;
| | - Giovanni Rossi
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, 07100 Sassari, Italy;
- UO Oncologia Medica, Ospedale Padre Antero Micone, 16153 Genova, Italy
| | - Silvia Morbelli
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.B.); (S.M.)
- Nuclear Medicine Unit, Department of Health Sciences, University of Genoa, 16132 Genoa, Italy
| | - Luca Guerra
- School of Medicine and Surgery, University of Milano Bicocca, 20900 Monza, Italy; (L.M.); (L.G.)
- Nuclear Medicine, ASST Monza San Gerardo Hospital, 20900 Monza, Italy;
| |
Collapse
|
60
|
Rundo F, Bersanelli M, Urzia V, Friedlaender A, Cantale O, Calcara G, Addeo A, Banna GL. Three-Dimensional Deep Noninvasive Radiomics for the Prediction of Disease Control in Patients With Metastatic Urothelial Carcinoma treated With Immunotherapy. Clin Genitourin Cancer 2021; 19:396-404. [PMID: 33849811 DOI: 10.1016/j.clgc.2021.03.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/09/2021] [Accepted: 03/13/2021] [Indexed: 01/15/2023]
Abstract
INTRODUCTION Immunotherapy is effective in a small percentage of patients with cancer and no reliable predictive biomarkers are currently available. Artificial Intelligence algorithms may automatically quantify radiologic characteristics associated with disease response to medical treatments. METHODS We investigated an innovative approach based on a 3-dimensional (3D) deep radiomics pipeline to classify visual features of chest-abdomen computed tomography (CT) scans with the aim of distinguishing disease control from progressive disease to immune checkpoint inhibitors (ICIs). Forty-two consecutive patients with metastatic urothelial cancer had progressed on first-line platinum-based chemotherapy and had baseline CT scans at immunotherapy initiation. The 3D-pipeline included self-learned visual features and a deep self-attention mechanism. According to the outcome to the ICIs, a 3D deep classifier semiautomatically categorized the most discriminative region of interest on the CT scans. RESULTS With a median follow-up of 13.3 months (95% CI, 11.1-15.6), the median overall survival was 8.5 months (95% CI, 3.1-13.8). According to disease response to immunotherapy, the median overall survival was 3.6 months (95% CI, 2.0-5.2) for patients with progressive disease; it was not yet reached for those with disease control. The predictive accuracy of the 3D-pipeline was 82.5% (sensitivity 96%; specificity, 60%). The addition of baseline clinical factors increased the accuracy to 92.5% by improving specificity to 87%; the accuracy of other architectures ranged from 72.5% to 90%. CONCLUSION Artificial Intelligence by 3D deep radiomics is a potential noninvasive biomarker for the prediction of disease control to ICIs in metastatic urothelial cancer and deserves validation in larger series.
Collapse
Affiliation(s)
| | - Melissa Bersanelli
- Medical Oncology Unit, Medicine and Surgery Department, University of Parma, Parma, Italy.
| | | | - Alex Friedlaender
- Oncology Department, Geneva University Hospital, Geneva, Switzerland
| | - Ornella Cantale
- Department of Experimental Oncology, Istituto Oncologico del Mediterraneo, Viagrande, Italy
| | - Giacomo Calcara
- Division of Medical Oncology and Department of Radiology, Cannizzaro Hospital, Catania, Italy
| | - Alfredo Addeo
- Oncology Department, Geneva University Hospital, Geneva, Switzerland
| | - Giuseppe Luigi Banna
- Division of Medical Oncology and Department of Radiology, Cannizzaro Hospital, Catania, Italy; Department of Oncology, Portsmouth Hospitals NHS Trust, Portsmouth, United Kingdom
| |
Collapse
|
61
|
Yin L, Zhou L, Xu R. Identification of Tumor Mutation Burden and Immune Infiltrates in Hepatocellular Carcinoma Based on Multi-Omics Analysis. Front Mol Biosci 2021; 7:599142. [PMID: 33681288 PMCID: PMC7928364 DOI: 10.3389/fmolb.2020.599142] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 12/21/2020] [Indexed: 12/16/2022] Open
Abstract
We aimed to explore the tumor mutational burden (TMB) and immune infiltration in HCC and investigate new biomarkers for immunotherapy. Transcriptome and gene mutation data were downloaded from the GDC portal, including 374 HCC samples and 50 matched normal samples. Furthermore, we divided the samples into high and low TMB groups, and analyzed the differential genes between them with GO, KEGG, and GSEA. Cibersort was used to assess the immune cell infiltration in the samples. Finally, univariate and multivariate Cox regression analyses were performed to identify differential genes related to TMB and immune infiltration, and a risk prediction model was constructed. We found 10 frequently mutated genes, including TP53, TTN, CTNNB1, MUC16, ALB, PCLO, MUC, APOB, RYR2, and ABCA. Pathway analysis indicated that these TMB-related differential genes were mainly enriched in PI3K-AKT. Cibersort analysis showed that memory B cells (p = 0.02), CD8+ T cells (p = 0.09), CD4+ memory activated T cells (p = 0.07), and neutrophils (p = 0.06) demonstrated a difference in immune infiltration between high and low TMB groups. On multivariate analysis, GABRA3 (p = 0.05), CECR7 (p < 0.001), TRIM16 (p = 0.003), and IL7R (p = 0.04) were associated with TMB and immune infiltration. The risk prediction model had an area under the curve (AUC) of 0.69, suggesting that patients with low risk had better survival outcomes. Our study demonstrated for the first time that CECR7, GABRA3, IL7R, and TRIM16L were associated with TMB and promoted antitumor immunity in HCC.
Collapse
Affiliation(s)
- Lu Yin
- Department of Pathology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liuzhi Zhou
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Rujun Xu
- Department of Pathology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
62
|
Kim SI, Cassella CR, Byrne KT. Tumor Burden and Immunotherapy: Impact on Immune Infiltration and Therapeutic Outcomes. Front Immunol 2021; 11:629722. [PMID: 33597954 PMCID: PMC7882695 DOI: 10.3389/fimmu.2020.629722] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 12/18/2020] [Indexed: 12/20/2022] Open
Abstract
Cancer immunotherapy has revolutionized the treatment landscape in medical oncology, but its efficacy has been variable across patients. Biomarkers to predict such differential response to immunotherapy include cytotoxic T lymphocyte infiltration, tumor mutational burden, and microsatellite instability. A growing number of studies also suggest that baseline tumor burden, or tumor size, predicts response to immunotherapy. In this review, we discuss the changes in immune profile and therapeutic responses that occur with increasing tumor size. We also overview therapeutic approaches to reduce tumor burden and favorably modulate the immune microenvironment of larger tumors.
Collapse
Affiliation(s)
- Samuel I Kim
- Program in Biochemistry, College of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, United States
| | - Christopher R Cassella
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Katelyn T Byrne
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|