1
|
Deng W, Wang Y, Wang J, Su Y, Li M, Qu K, Wang Y, Li M. Leveraging Vitamin C to Augment Nanoenabled Photothermal Immunotherapy. ACS NANO 2025; 19:12982-12995. [PMID: 40138545 DOI: 10.1021/acsnano.4c17080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Photothermal immunotherapy (PTI) is valuable for precise tumor targeting and immune activation. However, its efficacy is hindered by insufficient immune response, elevated antioxidant levels within tumor, and intrinsic tumor resistance mechanisms. This study introduces Vitamin C (VC), a widely available dietary nutrient, as an effective enhancer for PTI. High-dose VC induces oxidative imbalance in tumor cells, making them more susceptible to nanoenabled near-infrared-II photothermal therapy (NIR-II PTT) with the photosensitizer IR1080. The combination of VC and NIR-II PTT significantly amplifies antitumor immunity by upregulating CXCL16 expression and promoting CXCR6+ T cell infiltration. Clinical data reveal that higher CXCL16 and CXCR6 levels in human tumors correlate with improved survival and T cell infiltration, underscoring the translational potential of this approach. This study positions VC as a safe, accessible, and cost-effective dietary enhancer for PTI, reshaping the role of dietary nutrients in cancer therapy and offering a strategy for overcoming treatment resistance.
Collapse
Affiliation(s)
- Wuxian Deng
- Department of Radiology, the First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001 Anhui, China
- National Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027 Anhui, China
| | - Yiyuan Wang
- Department of Radiology, the First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001 Anhui, China
- National Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027 Anhui, China
| | - Junyu Wang
- National Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027 Anhui, China
| | - Yitan Su
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, 230601 Anhui, China
| | - Mingyang Li
- National Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027 Anhui, China
| | - Kun Qu
- National Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027 Anhui, China
| | - Yucai Wang
- Department of Radiology, the First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001 Anhui, China
- National Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027 Anhui, China
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, 230601 Anhui, China
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, 230027 Anhui, China
| | - Min Li
- Department of Radiology, the First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001 Anhui, China
- National Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027 Anhui, China
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, 230027 Anhui, China
| |
Collapse
|
2
|
Rademacher AF, Fadel HA, Pawloski JA, Ma M, Nkongchu KN, Lee IY, Ali AY. Laser Interstitial Thermal Therapy for Intra-Axial Brain Tumors: Everything the Neuroradiologist Should Know. AJNR Am J Neuroradiol 2025; 46:666-674. [PMID: 39572197 PMCID: PMC11979850 DOI: 10.3174/ajnr.a8427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/01/2024] [Indexed: 04/04/2025]
Abstract
Laser interstitial thermal therapy (LITT) is a minimally invasive cytoreductive treatment option for patients with intracranial tumors. Utilizing real-time MR thermometry, LITT delivers tailored, targeted, and permanent cytotoxic thermal injury to intra-axial pathology. As a minimally invasive and nonionizing treatment option proved to be an effective, less morbid, and more efficient alternative to surgery, the utility of LITT has rapidly expanded. Along with this growth comes the need for neurosurgeons and neuroradiologists to accurately assess the radiographic outcomes of LITT in a standardized, dependable, and longitudinal fashion. We present a comprehensive overview of the indications and mechanisms of action of LITT for intra-axial brain tumors as well as guidance on thorough pre-, intra-, and postoperative imaging assessments. Using detailed case examples describing the contemporary uses of LITT, we hope to provide a foundational understanding of LITT that will inform imaging assessment and guide accurate multi disciplinary tumor board discussion.
Collapse
Affiliation(s)
- Anneliese F Rademacher
- From the Department of Neurosurgery (A.F.R., H.A.F., J.A.P., I.Y.L.), Henry Ford Health, Detroit, Michigan
| | - Hassan A Fadel
- From the Department of Neurosurgery (A.F.R., H.A.F., J.A.P., I.Y.L.), Henry Ford Health, Detroit, Michigan
| | - Jacob A Pawloski
- From the Department of Neurosurgery (A.F.R., H.A.F., J.A.P., I.Y.L.), Henry Ford Health, Detroit, Michigan
| | - Mia Ma
- Department of Radiology (M.M., K.N.N., A.Y.A.), Henry Ford Health, Detroit, Michigan
| | - Ken N Nkongchu
- Department of Radiology (M.M., K.N.N., A.Y.A.), Henry Ford Health, Detroit, Michigan
| | - Ian Y Lee
- From the Department of Neurosurgery (A.F.R., H.A.F., J.A.P., I.Y.L.), Henry Ford Health, Detroit, Michigan
| | - Arafat Y Ali
- Department of Radiology (M.M., K.N.N., A.Y.A.), Henry Ford Health, Detroit, Michigan
| |
Collapse
|
3
|
Almeida T, Reyes JS, Scanlon SE, Frederico SC, Huq S, Hadjipanayis CG. Laser interstitial thermal therapy for recurrent brain metastases following stereotactic radiosurgery: a systematic review. Expert Rev Neurother 2025; 25:465-475. [PMID: 39991830 DOI: 10.1080/14737175.2025.2471551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025]
Abstract
INTRODUCTION Laser Interstitial Thermal Therapy (LITT) has emerged as a promising minimally invasive treatment option for recurrent brain metastases, especially for patients who have previously undergone Stereotactic Radiosurgery (SRS). Despite its growing use, additional research is needed to explore its utility and safety in this population. METHODS This systematic review evaluates the literature on LITT for recurrent brain metastases post-SRS, including patient selection, treatment protocols, outcomes, and complications. The review was registered with PROSPERO (CRD42024577899) and followed the PRISMA 2020 guidelines. RESULTS Seventeen studies, encompassing 694 patients, met our inclusion criteria. The median overall survival (OS) across these studies was 14.4 months with an average 12-month OS of 60.1%. Notably, median OS was 5.9 months after LITT alone and 12.7 months when LITT was followed by adjuvant SRS. The pooled local control rate was 75.6% at 6 months and 72.2% at 12 months. CONCLUSION LITT is a minimally invasive treatment option that has shown potential in managing recurrent brain metastases after prior SRS, particularly when biopsy is required to differentiate between tumor recurrence and radiation necrosis. However, data on the comparative efficacy and cost-effectiveness of LITT versus alternative treatments such as repeat SRS or craniotomy remain limited.
Collapse
Affiliation(s)
- Timoteo Almeida
- Center for Image-Guided Neurosurgery, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, USA
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Radiation Oncology, University of Miami, Miami, FL, USA
| | - Jheremy S Reyes
- Center for Image-Guided Neurosurgery, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, USA
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sydney E Scanlon
- Center for Image-Guided Neurosurgery, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, USA
| | - Stephen C Frederico
- Center for Image-Guided Neurosurgery, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, USA
| | - Sakibul Huq
- Center for Image-Guided Neurosurgery, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, USA
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Costas G Hadjipanayis
- Center for Image-Guided Neurosurgery, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, USA
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
4
|
Line TA, Elkinbard IS, Purger DA, Buch VP. Editorial for Brain Sciences Special Issue "Advances in Restorative Neurotherapeutic Technologies". Brain Sci 2025; 15:273. [PMID: 40149794 PMCID: PMC11940112 DOI: 10.3390/brainsci15030273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 02/13/2025] [Indexed: 03/29/2025] Open
Abstract
From Ramon y Cajal and Golgi's histological techniques to single-cell RNA sequencing, technological innovations have long driven progress in neuroscience [...].
Collapse
Affiliation(s)
- Trenton A. Line
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | | | - David A. Purger
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Vivek P. Buch
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA;
| |
Collapse
|
5
|
Sharma E, Sahin OK, Łajczak P, Rajab N, Ahmed AR, Silva YP, Bakhsh A, Chatterjee A, Raake M, Fagundes W. Synergistic effects of laser interstitial thermal therapy (LITT) and immunotherapy for brain tumor recurrence: A systematic review and meta-analysis. Neurochirurgie 2025; 71:101629. [PMID: 39756615 DOI: 10.1016/j.neuchi.2025.101629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/11/2024] [Accepted: 12/21/2024] [Indexed: 01/07/2025]
Abstract
PURPOSE The confluence of laser interstitial thermal therapy (LITT) with immunotherapeutic approaches represents a promising option for managing recurrent brain lesions. However, the potential synergy between these modalities is still unclear. This meta-analysis examines the literature to elucidate the adverse effects and overall survival associated with this combination in treating recurrent brain metastases and glioblastoma. METHODS Systematic searches were performed on PubMed, Embase, and Web of Science databases. Inclusion criteria comprised studies investigating the combined utilization of LITT with immunotherapy, among adult patients diagnosed with recurrent brain metastases and recurrent glioblastoma. Our analysis, using a random-effects model, pooled Overall Survival (OS) and Adverse events (AEs) from all the included studies. RESULTS We analyzed 162 patients from one RCT and three non-randomized studies. The pooled analysis of all patients revealed a median OS of 12.8 months (95% CI = 8.31-17.31; p < 0.01) with the combined treatment of LITT and immunotherapy. Similarly, approximately 6% of patients experienced AEs (95% CI = -0.01-0.11; p = 0.03). Subgroup analysis further demonstrated that among patients with recurrent glioma, the combined treatment showed pooled OS of 11 months (95% CI = 7.13-16.62; p < 0.01), while AEs were observed in 4% of patients (95% CI = -0.02-0.10; p = 0.21). CONCLUSION This meta-analysis showed a potentially comparable safety profile and overall survival to conventional treatment modalities. Further research is warranted to test differences in the incidence of AEs and OS from LITT with immunotherapy versus a control.
Collapse
Affiliation(s)
- Eshita Sharma
- David Geffen School of Medicine at UCLA, Los Angeles, United States.
| | | | | | - Numa Rajab
- Sulaiman AlRajhi University, Saudi Arabia
| | | | | | | | | | | | - Walter Fagundes
- Department of Neurosurgery, Federal University of Espírito Santo, Vitória, Brazil
| |
Collapse
|
6
|
Haskell-Mendoza AP, Gonzalez AT, Reason EH, Flusche AM, Chongsathidkiet P, Wachsmuth LP, Goodwin CR, Fecci PE. The LITT Fit in neuro-oncology: indications, imaging, and adjunctive therapies. J Neurooncol 2025; 172:1-11. [PMID: 39585599 DOI: 10.1007/s11060-024-04894-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 11/18/2024] [Indexed: 11/26/2024]
Abstract
PURPOSE There is an unmet need for new treatments for many central nervous system tumors. An expanding body of research supports the use of laser interstitial thermal therapy (LITT) in the treatment of gliomas, recurrent brain metastases, and radiation necrosis. METHODS In this review, we highlight emerging indications for LITT, including its use adjacent to eloquent structures, in the posterior fossa, and for meningioma and tumors of the vertebral column. We conclude by providing an overview of current research into post-LITT response assessment and adjunctive therapies. RESULTS Evidence has continued to accumulate regarding the safety of LITT in locations as varied as the motor cortex, posterior fossa, and vertebral column, as well as for novel pathologies such as meningioma. Regardless of disease histology, most patients leave the hospital within 12-48 h of LITT and can rapidly return to systemic and radiation therapies. Emerging data has allowed for a characterization of post-LITT imaging findings, and receipt of LITT should not preclude subsequent clinical trial enrollment, especially as hyperthermia modulates blood-brain barrier permeability and may synergize with immunotherapies. CONCLUSION As LITT is incorporated into neurosurgical oncology practice, novel use cases will continue to emerge. Given that laser ablation is associated with shortened length of stay and decreased debility relative to open resection, development of radiographic response assessment criteria for LITT-treated lesions is urgently needed so that patients may more rapidly receive definitive management or proceed to clinical trial enrollment. Prospective evaluation of LITT and adjunctive combination therapies is ongoing.
Collapse
Affiliation(s)
- Aden P Haskell-Mendoza
- The Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
| | - Ariel T Gonzalez
- The Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
| | - Ellery H Reason
- The Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
| | - Ann Marie Flusche
- The Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
| | - Pakawat Chongsathidkiet
- The Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
| | - Lucas P Wachsmuth
- The Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
| | - C Rory Goodwin
- Duke Center for Brain and Spine Metastasis, Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
| | - Peter E Fecci
- The Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA.
- Duke Center for Brain and Spine Metastasis, Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
7
|
Zhao X, Li C, Wang K, Zhang Y, Wan H, Yang P, Zhou D. MR‑guided laser interstitial thermal therapy followed by early application of temozolomide for recurrent IDH-wildtype glioblastomas: preliminary results from a prospective study. Neurosurg Rev 2025; 48:254. [PMID: 39971799 DOI: 10.1007/s10143-025-03402-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 01/19/2025] [Accepted: 02/09/2025] [Indexed: 02/21/2025]
Abstract
This study aims to evaluate the safety, tolerability, and preliminary efficacy of combining laser interstitial thermal therapy (LITT) with early administration of temozolomide (TMZ) in patients with recurrent glioblastoma (rGBM). Ten patients with rGBM were enrolled. Following the LITT procedure, TMZ was administered at a dose of 75 mg/m2/day during the early-TMZ phase for three weeks. After a 7-day interval, TMZ was given according to the standard dosage scheme for 6 cycles. Adverse events and complications encountered were documented. Regular follow-up assessments were conducted to evaluate both patient performance status and tumor progression. All patients demonstrated good tolerance to LITT, with six out of ten achieving an ablation rate above 90%, and only one patient had an ablation rate below 70%. Oral administration of TMZ was well-tolerated by all patients during the early-TMZ phase. Mild headache was the most common adverse event (3/10), and only one severe adverse event occurred. At a 6-month follow-up post-LITT, tumor progression was observed in five patients; noneof the patients reached survival endpoints. This preliminary report substantiates the favorable tolerability of early application of TMZ in combination with LITT. The safety profile was found to be acceptable, and the initial efficacy results were promising. Future studies should explore the potential of LITT combination therapy in greater detail and with larger patient samples.
Collapse
Affiliation(s)
- Xuzhe Zhao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No.119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, China
| | - Chao Li
- Department of Neurosurgery, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Kai Wang
- Department of Nuclear Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yutao Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No.119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, China
| | - Haibin Wan
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No.119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, China
| | - Pei Yang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No.119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, China
| | - Dabiao Zhou
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No.119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, China.
| |
Collapse
|
8
|
Rodriguez B, Rivera D, Zhang JY, Brown C, Young T, Williams T, Kallos J, Huq S, Hadjpanayis C. Innovations in intraoperative therapies in neurosurgical oncology: a narrative review. J Neurooncol 2025; 171:549-557. [PMID: 39546148 DOI: 10.1007/s11060-024-04882-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 11/04/2024] [Indexed: 11/17/2024]
Abstract
PURPOSE High-grade gliomas (HGG) represent the most aggressive primary brain tumors in adults, characterized by high recurrence rates due to incomplete resection. This review explores the effectiveness of emerging intraoperative therapies that may extend survival by targeting residual tumor cells. The main research question addressed is: What recent intraoperative techniques show promise for complementing surgical resection in HGG treatment? METHODS A comprehensive literature review was conducted, examining recent studies on intraoperative therapeutic modalities that support surgical resection of HGG. Techniques reviewed include laser interstitial thermal therapy (LITT), intraoperative brachytherapy, photodynamic therapy (PDT), sonodynamic therapy (SDT), and focused ultrasound (FUS). Each modality was evaluated based on clinical application, evidence of effectiveness, and potential for integration into standard HGG treatment protocols. RESULTS Findings indicate that these therapies offer distinct mechanisms to target residual tumor cells: LITT provides localized thermal ablation; intraoperative brachytherapy delivers sustained radiation; PDT and SDT activate cytotoxic agents in tumor cells; and FUS enables precise energy delivery. Each method has shown varying levels of clinical success, with PDT and LITT currently more widely implemented, while SDT and FUS are promising but under investigation. CONCLUSION Intraoperative therapies hold potential to improve surgical outcomes for HGG by reducing residual tumor burden. While further clinical studies are needed to optimize these techniques, early evidence supports their potential to enhance the effectiveness of surgical resection and improve patient survival in HGG management.
Collapse
Affiliation(s)
- Benjamin Rodriguez
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Sinai BioDesign, Department of Neurosurgery, Mount Sinai, New York, NY, USA
| | - Daniel Rivera
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jack Y Zhang
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Cole Brown
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tirone Young
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Sinai BioDesign, Department of Neurosurgery, Mount Sinai, New York, NY, USA
| | - Tyree Williams
- Sinai BioDesign, Department of Neurosurgery, Mount Sinai, New York, NY, USA
- Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Justiss Kallos
- Department of Neurological Surgery, UPMC, Pittsburgh, PA, USA
| | - Sakibul Huq
- Department of Neurological Surgery, UPMC, Pittsburgh, PA, USA
| | - Constantinos Hadjpanayis
- Department of Neurological Surgery, UPMC, Pittsburgh, PA, USA.
- Brain Tumor Nanotechnology Laboratory, UPMC Hillman Cancer Center Pittsburgh, 200 Lothrop Street, Suite F-158, Pittsburgh, PA, 15213, USA.
- Center for Image-Guided Neurosurgery, Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
9
|
McGrath K, Frain M, Hey G, Rahman M. Complications following laser interstitial thermal therapy: a review. Neurochirurgie 2025; 71:101604. [PMID: 39413572 DOI: 10.1016/j.neuchi.2024.101604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/29/2024] [Accepted: 10/05/2024] [Indexed: 10/18/2024]
Abstract
Laser interstitial thermal therapy (LITT) is being performed more frequently for various lesions within neurosurgery, including epileptic foci, vascular malformations, and tumors. Though this technique generally has an excellent safety profile, it is important to be aware of potential complications. Thermal ablation of tissue leads to disruption of the blood brain barrier as well as an inflammatory response both of which cause the majority of complications from LITT. The most common complications of LITT include cerebral edema, focal neurologic deficits, and intracranial hemorrhage. Few studies have identified factors predicting development of these complications, but many of these are transient and resolve without intervention. Modifications to LITT technique that allows better visualization of patient anatomy along the tract, such as fusing vascular imaging with intraoperative MRI, reduce the risk of complications.
Collapse
Affiliation(s)
- Kyle McGrath
- College of Medicine, University of Florida, Gainesville, FL, United States.
| | - Matthew Frain
- Department of Medical Physics, University of Florida, Gainesville, FL, United States; Brain Tumor Immunotherapy Program, University of Florida, Gainesville, FL, United States
| | - Grace Hey
- College of Medicine, University of Florida, Gainesville, FL, United States
| | - Maryam Rahman
- Brain Tumor Immunotherapy Program, University of Florida, Gainesville, FL, United States; Lillian S Wells Department of Neurosurgery, University of Florida, Gainesville, FL, United States
| |
Collapse
|
10
|
Yu JS, Meade SM, Zhao R, Wei W, Dashora H, Prayson R, Grabowski MM, Stevens G, Lobbous M, Murphy ES, Suh JH, Chao ST, Barnett GH, Peereboom D, Ahluwalia MS, Mohammadi AM. Expedited chemoradiation after laser interstitial thermal therapy (LITT) is feasible and safe in patients with newly diagnosed glioblastoma. Neurooncol Adv 2025; 7:vdaf038. [PMID: 40276375 PMCID: PMC12019959 DOI: 10.1093/noajnl/vdaf038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2025] Open
Abstract
Background High-grade gliomas (HGG) are incurable primary brain tumors. Laser interstitial thermal therapy (LITT) has emerged as an alternative to surgery for select patients. Hyperthermia can improve the efficacy of radiation and chemotherapy. Shortening the time between LITT and chemoradiation may maximize their biological and clinical benefits. This trial evaluated the safety and feasibility of expediting chemoradiation after biopsy and LITT in patients with newly diagnosed HGG. Methods Patients with suspected HGG were enrolled. Those with pathologic confirmation of HGG and deemed appropriate candidates for LITT and chemoradiation were considered evaluable. Participants underwent 6 weeks of adjuvant chemoradiation initiated within 7 days of LITT. Endpoints were assessed until the completion of radiation and included the occurrence of wound dehiscence; new, treatment-refractory seizures; cerebral edema; and completion of planned radiotherapy. Results Thirteen patients with suspected HGG were enrolled, and ten were considered evaluable. All 10 patients were diagnosed with glioblastoma (GBM, IDHwt). Three patients were deemed unevaluable: 2 patients with other CNS tumors and one GBM patient who developed grade 4 postoperative edema. Of 10 evaluable patients, the median age was 60.2 years (IQR: 51.0, 69.4), and median preoperative KPS was 90 (IQR: 90, 80). The median time between LITT and the initiation of chemoradiation was 7 days. There were no occurrences of significant protocol-related adverse events. Conclusions Accelerated initiation of chemoradiation after biopsy and LITT is safe and feasible for patients with newly diagnosed GBM. A larger study is needed to assess potential synergy of hyperthermia and chemoradiation to improve survival.
Collapse
Affiliation(s)
- Jennifer S Yu
- Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Cancer Biology, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Radiation Oncology, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Seth M Meade
- Department of Neurosurgery, Cleveland Clinic, Cleveland, Ohio, USA
- Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Ran Zhao
- Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio, USA
| | - Wei Wei
- Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio, USA
| | - Himanshu Dashora
- Case Western Reserve University School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Cancer Biology, Cleveland Clinic, Cleveland, Ohio, USA
| | - Richard Prayson
- Department of Pathology, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Matthew M Grabowski
- Department of Neurosurgery, Cleveland Clinic, Cleveland, Ohio, USA
- Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Glen Stevens
- Department of Neurology, Cleveland Clinic, Cleveland, Ohio, USA
- Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
| | - Mina Lobbous
- Department of Neurology, Cleveland Clinic, Cleveland, Ohio, USA
- Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Erin S Murphy
- Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Radiation Oncology, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - John H Suh
- Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Radiation Oncology, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Samuel T Chao
- Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Radiation Oncology, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Gene H Barnett
- Department of Neurosurgery, Cleveland Clinic, Cleveland, Ohio, USA
- Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - David Peereboom
- Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Manmeet S Ahluwalia
- Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
| | - Alireza M Mohammadi
- Department of Neurosurgery, Cleveland Clinic, Cleveland, Ohio, USA
- Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
11
|
Xie J, Li D, Niu S, Sheng Y, Shen R, He Y, Xu C, Zhang Y, Wang T, Xue Y. Nano-Titanium Oxide-Coated Carbon Nanotubes for Photothermal Therapy in the Treatment of Colorectal Cancer. Adv Healthc Mater 2024; 13:e2401009. [PMID: 38885692 DOI: 10.1002/adhm.202401009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 06/11/2024] [Indexed: 06/20/2024]
Abstract
Carbon nanotubes (CNTs) display good potential in tumor photothermal therapy (PTT). In this study, it is aimed to investigate the therapeutic potential of nano-titanium oxide-coated multi-walled carbon nanotubes (MCNTs) against colorectal cancer (CRC). First, TiO2 nanosheets are modified on the surface of MCNTs to obtain nano-TiO2-coated MCNTs. Next, cell compatibility validation is conducted on nano-TiO2-coated MCNTs, and it is found that nano-TiO2-coated MCNTs are safe within a certain concentration range (0-200 µg mL⁻1). Interestingly, nano-TiO2-coated MCNTs display a good killing effect in CRC cells under near-infrared (NIR) laser irradiation. Subsequently, nano-TiO2-coated MCNTs markedly promote the proapoptotic effects of NIR laser irradiation and significantly inhibit the expression of cell cycle proteins CCNA1 and CCND1 in CRC cells under NIR laser irradiation, which indicates that nano-TiO2-coated MCNTs exert anti-CRC effects under NIR laser irradiation by regulating cell apoptosis and cell cycle. Furthermore, nano-TiO2-coated MCNTs accelerate inhibitory effects on the AKT signaling pathway under NIR laser irradiation. Finally, a cell line-derived xenograft model is established, and the results showed that nano-TiO2-coated MCNTs significantly exhibit superior tumor-killing ability under NIR laser irradiation in vivo. Collectively, these results demonstrate that nano-TiO2-coated MCNTs with NIR laser irradiation may serve as an effective strategy for the treatment of CRC.
Collapse
Affiliation(s)
- Jun Xie
- Department of Pediatric Internal Medicine, Affiliated Children's Hospital of Jiangnan University (Wuxi Children's Hospital), Wuxi, Jiangsu, 214023, China
| | - Da Li
- Department of General Surgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Sen Niu
- Department of General Surgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Yufan Sheng
- Department of General Surgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Renhui Shen
- Department of General Surgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Yiding He
- Department of General Surgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Chenhao Xu
- Department of General Surgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Ye Zhang
- Department of General Surgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Tong Wang
- Department of General Surgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Yuzheng Xue
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214122, China
| |
Collapse
|
12
|
Nielsen SH, Rasmussen R. MR-guided laser interstitial thermal therapy in the treatment of brain tumors and epilepsy. Acta Neurochir (Wien) 2024; 166:344. [PMID: 39167226 DOI: 10.1007/s00701-024-06238-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 08/14/2024] [Indexed: 08/23/2024]
Abstract
MR-guided Laser Interstitial Thermal Therapy (MRgLITT) is a minimally invasive neurosurgical technique increasingly used for the treatment of drug-resistant epilepsy and brain tumors. Utilizing near-infrared light energy delivery guided by real-time MRI thermometry, MRgLITT enables precise ablation of targeted brain tissues, resulting in limited corridor-related morbidity and expedited postoperative recovery. Since receiving CE marking in 2018, the adoption of MRgLITT has expanded to more than 40 neurosurgical centers across Europe. In epilepsy treatment, MRgLITT can be applied to various types of focal lesional epilepsy, including mesial temporal lobe epilepsy, hypothalamic hamartoma, focal cortical dysplasias, periventricular heterotopias, cavernous malformations, dysembryoplastic neuroepithelial tumors (DNET), low-grade gliomas, tuberous sclerosis, and in disconnective surgeries. In neuro-oncology, MRgLITT is used for treating newly diagnosed and recurrent primary brain tumors, brain metastases, and radiation necrosis. This comprehensive review presents an overview of the current evidence and technical considerations for the use of MRgLITT in treating various pathologies associated with drug-resistant epilepsy and brain tumors.
Collapse
Affiliation(s)
- Silas Haahr Nielsen
- Department of Neurosurgery, Copenhagen University Hospital, Copenhagen, Denmark.
| | - Rune Rasmussen
- Department of Neurosurgery, Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
13
|
Lu YJ, Vayalakkara RK, Dash BS, Hu SH, Premji TP, Wu CY, Shen YJ, Chen JP. Immunomodulatory R848-Loaded Anti-PD-L1-Conjugated Reduced Graphene Oxide Quantum Dots for Photothermal Immunotherapy of Glioblastoma. Pharmaceutics 2024; 16:1064. [PMID: 39204409 PMCID: PMC11358977 DOI: 10.3390/pharmaceutics16081064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 07/30/2024] [Accepted: 08/09/2024] [Indexed: 09/04/2024] Open
Abstract
Glioblastoma multiforme (GBM) is the most severe form of brain cancer and presents unique challenges to developing novel treatments due to its immunosuppressive milieu where receptors like programmed death ligand 1 (PD-L1) are frequently elevated to prevent an effective anti-tumor immune response. To potentially shift the GBM environment from being immunosuppressive to immune-enhancing, we engineered a novel nanovehicle from reduced graphene oxide quantum dot (rGOQD), which are loaded with the immunomodulatory drug resiquimod (R848) and conjugated with an anti-PD-L1 antibody (aPD-L1). The immunomodulatory rGOQD/R8/aPDL1 nanoparticles can actively target the PD-L1 on the surface of ALTS1C1 murine glioblastoma cells and release R848 to enhance the T-cell-driven anti-tumor response. From in vitro experiments, the PD-L1-mediated intracellular uptake and the rGOQD-induced photothermal response after irradiation with near-infrared laser light led to the death of cancer cells and the release of damage-associated molecular patterns (DAMPs). The combinational effect of R848 and released DAMPs synergistically produces antigens to activate dendritic cells, which can prime T lymphocytes to infiltrate the tumor in vivo. As a result, T cells effectively target and attack the PD-L1-suppressed glioma cells and foster a robust photothermal therapy elicited anti-tumor immune response from a syngeneic mouse model of GBM with subcutaneously implanted ALTS1C1 cells.
Collapse
Affiliation(s)
- Yu-Jen Lu
- Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, Kwei-San, Taoyuan 33305, Taiwan; (Y.-J.L.); (R.K.V.)
- College of Medicine, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
| | - Reesha Kakkadavath Vayalakkara
- Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, Kwei-San, Taoyuan 33305, Taiwan; (Y.-J.L.); (R.K.V.)
| | - Banendu Sunder Dash
- Department of Chemical and Materials and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan; (B.S.D.)
| | - Shang-Hsiu Hu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300044, Taiwan
| | - Thejas Pandaraparambil Premji
- Department of Chemical and Materials and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan; (B.S.D.)
| | - Chun-Yuan Wu
- Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, Kwei-San, Taoyuan 33305, Taiwan; (Y.-J.L.); (R.K.V.)
| | - Yang-Jin Shen
- Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, Kwei-San, Taoyuan 33305, Taiwan; (Y.-J.L.); (R.K.V.)
| | - Jyh-Ping Chen
- Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, Kwei-San, Taoyuan 33305, Taiwan; (Y.-J.L.); (R.K.V.)
- Department of Chemical and Materials and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan; (B.S.D.)
- Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33302, Taiwan
- Department of Materials Engineering, Ming Chi University of Technology, Tai-Shan, New Taipei City 24301, Taiwan
| |
Collapse
|
14
|
Liang T, Gu L, Kang X, Li J, Song Y, Wang Y, Ma W. Programmed cell death disrupts inflammatory tumor microenvironment (TME) and promotes glioblastoma evolution. Cell Commun Signal 2024; 22:333. [PMID: 38890642 PMCID: PMC11184850 DOI: 10.1186/s12964-024-01602-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 04/01/2024] [Indexed: 06/20/2024] Open
Abstract
Glioblastoma (GBM) is the most common malignant brain tumor and has a dismal prognosis even under the current first-line treatment, with a 5-year survival rate less than 7%. Therefore, it is important to understand the mechanism of treatment resistance and develop new anti-tumor strategies. Induction of programmed cell death (PCD) has become a promising anti-tumor strategy, but its effectiveness in treating GBM remains controversial. On the one hand, PCD triggers tumor cell death and then release mediators to draw in immune cells, creating a pro-inflammatory tumor microenvironment (TME). One the other hand, mounting evidence suggests that PCD and inflammatory TME will force tumor cells to evolve under survival stress, leading to tumor recurrence. The purpose of this review is to summarize the role of PCD and inflammatory TME in the tumor evolution of GBM and promising methods to overcome tumor evolution.
Collapse
Affiliation(s)
- Tingyu Liang
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT Alliance, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Lingui Gu
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT Alliance, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Xiaoman Kang
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT Alliance, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- '4+4' Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Junlin Li
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT Alliance, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- Eight-year Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yixuan Song
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT Alliance, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- Eight-year Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yu Wang
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT Alliance, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Wenbin Ma
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT Alliance, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
15
|
Haskell-Mendoza AP, Srinivasan ES, Vo-Dinh T, Fecci PE. Leveraging gold nanostars for precision laser interstitial thermal therapy. Oncotarget 2024; 15:389-391. [PMID: 38874556 DOI: 10.18632/oncotarget.28592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2024] Open
|
16
|
Pandey A, Chandla A, Mekonnen M, Hovis GEA, Teton ZE, Patel KS, Everson RG, Wadehra M, Yang I. Safety and Efficacy of Laser Interstitial Thermal Therapy as Upfront Therapy in Primary Glioblastoma and IDH-Mutant Astrocytoma: A Meta-Analysis. Cancers (Basel) 2024; 16:2131. [PMID: 38893250 PMCID: PMC11171930 DOI: 10.3390/cancers16112131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 05/24/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
Although primary studies have reported the safety and efficacy of LITT as a primary treatment in glioma, they are limited by sample sizes and institutional variation in stereotactic parameters such as temperature and laser power. The current literature has yet to provide pooled statistics on outcomes solely for primary brain tumors according to the 2021 WHO Classification of Tumors of the Central Nervous System (WHO CNS5). In the present study, we identify recent articles on primary CNS neoplasms treated with LITT without prior intervention, focusing on relationships with molecular profile, PFS, and OS. This meta-analysis includes the extraction of data from primary sources across four databases using the Covidence systematic review manager. The pooled data suggest LITT may be a safe primary management option with tumor ablation rates of 94.8% and 84.6% in IDH-wildtype glioblastoma multiforme (GBM) and IDH-mutant astrocytoma, respectively. For IDH-wildtype GBM, the pooled PFS and OS were 5.0 and 9.0 months, respectively. Similar to rates reported in the prior literature, the neurologic and non-neurologic complication rates for IDH-wildtype GBM were 10.3% and 4.8%, respectively. The neurologic and non-neurologic complication rates were somewhat higher in the IDH-mutant astrocytoma cohort at 33% and 8.3%, likely due to a smaller cohort size.
Collapse
Affiliation(s)
- Aryan Pandey
- Department of Neurosurgery, University of California Los Angeles (UCLA), Los Angeles, CA 90095, USA (M.M.)
| | - Anubhav Chandla
- Department of Neurosurgery, University of California Los Angeles (UCLA), Los Angeles, CA 90095, USA (M.M.)
| | - Mahlet Mekonnen
- Department of Neurosurgery, University of California Los Angeles (UCLA), Los Angeles, CA 90095, USA (M.M.)
| | - Gabrielle E. A. Hovis
- Department of Neurosurgery, University of California Los Angeles (UCLA), Los Angeles, CA 90095, USA (M.M.)
| | - Zoe E. Teton
- Department of Neurosurgery, University of California Los Angeles (UCLA), Los Angeles, CA 90095, USA (M.M.)
- Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Kunal S. Patel
- Department of Neurosurgery, University of California Los Angeles (UCLA), Los Angeles, CA 90095, USA (M.M.)
- Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095, USA
| | - Richard G. Everson
- Department of Neurosurgery, University of California Los Angeles (UCLA), Los Angeles, CA 90095, USA (M.M.)
- Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095, USA
- Department of Radiation Oncology, University of California Los Angeles (UCLA), Los Angeles, CA 90095, USA
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
- Ronald Reagan UCLA Medical Center, Los Angeles, CA 90095, USA
| | - Madhuri Wadehra
- Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095, USA
- Department of Pathology and Laboratory Medicine, University of California Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Isaac Yang
- Department of Neurosurgery, University of California Los Angeles (UCLA), Los Angeles, CA 90095, USA (M.M.)
- Harbor-UCLA Medical Center, Torrance, CA 90502, USA
- Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095, USA
- Department of Radiation Oncology, University of California Los Angeles (UCLA), Los Angeles, CA 90095, USA
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
- Ronald Reagan UCLA Medical Center, Los Angeles, CA 90095, USA
- Department of Head and Neck Surgery, University of California Los Angeles (UCLA), Los Angeles, CA 90095, USA
| |
Collapse
|
17
|
Haskell-Mendoza AP, Reason EH, Gonzalez AT, Jackson JD, Sankey EW, Srinivasan ES, Herndon JE, Fecci PE, Calabrese E. Automated segmentation of ablated lesions using deep convolutional neural networks: A basis for response assessment following laser interstitial thermal therapy. Neuro Oncol 2024; 26:1152-1162. [PMID: 38170451 PMCID: PMC11145442 DOI: 10.1093/neuonc/noad261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Laser interstitial thermal therapy (LITT) of intracranial tumors or radiation necrosis enables tissue diagnosis, cytoreduction, and rapid return to systemic therapies. Ablated tissue remains in situ, resulting in characteristic post-LITT edema associated with transient clinical worsening and complicating post-LITT response assessment. METHODS All patients receiving LITT at a single center for tumors or radiation necrosis from 2015 to 2023 with ≥9 months of MRI follow-up were included. An nnU-Net segmentation model was trained to automatically segment contrast-enhancing lesion volume (CeLV) of LITT-treated lesions on T1-weighted images. Response assessment was performed using volumetric measurements. RESULTS Three hundred and eighty four unique MRI exams of 61 LITT-treated lesions and 6 control cases of medically managed radiation necrosis were analyzed. Automated segmentation was accurate in 367/384 (95.6%) images. CeLV increased to a median of 68.3% (IQR 35.1-109.2%) from baseline at 1-3 months from LITT (P = 0.0012) and returned to baseline thereafter. Overall survival (OS) for LITT-treated patients was 39.1 (9.2-93.4) months. Lesion expansion above 40% from volumetric nadir or baseline was considered volumetric progression. Twenty-one of 56 (37.5%) patients experienced progression for a volumetric progression-free survival of 21.4 (6.0-93.4) months. Patients with volumetric progression had worse OS (17.3 vs 62.1 months, P = 0.0015). CONCLUSIONS Post-LITT CeLV expansion is quantifiable and resolves within 6 months of LITT. Development of response assessment criteria for LITT-treated lesions is feasible and should be considered for clinical trials. Automated lesion segmentation could speed the adoption of volumetric response criteria in clinical practice.
Collapse
Affiliation(s)
| | - Ellery H Reason
- Duke University School of Medicine, Durham, North Carolina, USA
| | | | - Joshua D Jackson
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Eric W Sankey
- Department of Neurosurgery, Piedmont Athens Regional Medical Center, Athens, Georgia, USA
| | - Ethan S Srinivasan
- Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - James E Herndon
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Peter E Fecci
- The Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Evan Calabrese
- Department of Radiology, Division of Neuroradiology, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
18
|
Martines-Arano H, Valdivia-Flores A, Castillo-Cruz J, García-Pérez BE, Torres-Torres C. Spatially modulated ablation driven by chaotic attractors in human lung epithelial cancer cells. Biomed Phys Eng Express 2024; 10:035041. [PMID: 38569484 DOI: 10.1088/2057-1976/ad39f1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 04/03/2024] [Indexed: 04/05/2024]
Abstract
A significant modification in photoinduced energy transfer in cancer cells is reported by the assistance of a dynamic modulation of the beam size of laser irradiation. Human lung epithelial cancer cells in monolayer form were studied. In contrast to the quantum and thermal ablation effect promoted by a standard focused Gaussian beam, a spatially modulated beam can caused around 15% of decrease in the ablation threshold and formation of a ring-shaped distribution of the photothermal transfer effect. Optical irradiation was conducted in A549 cells by a 532 nm single-beam emerging from a Nd:YVO4 system. Ablation effects derived from spatially modulated convergent waves were controlled by an electrically focus-tunable lens. The proposed chaotic behavior of the spatial modulation followed an Arneodo chaotic oscillator. Fractional dynamic thermal transport was analyzed in order to describe photoenergy in propagation through the samples. Immediate applications of chaos theory for developing phototechnology devices driving biological functions or phototherapy treatments can be considered.
Collapse
Affiliation(s)
- Hilario Martines-Arano
- Facultad de Ciencias de la Electrónica, Benemérita Universidad Autónoma de Puebla, 72592, Puebla, Puebla, Mexico
| | - Alejandra Valdivia-Flores
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, 11340, Mexico
| | - Juan Castillo-Cruz
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, 11340, Mexico
| | - Blanca Estela García-Pérez
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, 11340, Mexico
| | - Carlos Torres-Torres
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Ingeniería Mecánica y Eléctrica Unidad Zacatenco, Instituto Politécnico Nacional, Ciudad de México, 07738, Mexico
| |
Collapse
|
19
|
Obrador E, Moreno-Murciano P, Oriol-Caballo M, López-Blanch R, Pineda B, Gutiérrez-Arroyo JL, Loras A, Gonzalez-Bonet LG, Martinez-Cadenas C, Estrela JM, Marqués-Torrejón MÁ. Glioblastoma Therapy: Past, Present and Future. Int J Mol Sci 2024; 25:2529. [PMID: 38473776 PMCID: PMC10931797 DOI: 10.3390/ijms25052529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 02/10/2024] [Accepted: 02/16/2024] [Indexed: 03/14/2024] Open
Abstract
Glioblastoma (GB) stands out as the most prevalent and lethal form of brain cancer. Although great efforts have been made by clinicians and researchers, no significant improvement in survival has been achieved since the Stupp protocol became the standard of care (SOC) in 2005. Despite multimodality treatments, recurrence is almost universal with survival rates under 2 years after diagnosis. Here, we discuss the recent progress in our understanding of GB pathophysiology, in particular, the importance of glioma stem cells (GSCs), the tumor microenvironment conditions, and epigenetic mechanisms involved in GB growth, aggressiveness and recurrence. The discussion on therapeutic strategies first covers the SOC treatment and targeted therapies that have been shown to interfere with different signaling pathways (pRB/CDK4/RB1/P16ink4, TP53/MDM2/P14arf, PI3k/Akt-PTEN, RAS/RAF/MEK, PARP) involved in GB tumorigenesis, pathophysiology, and treatment resistance acquisition. Below, we analyze several immunotherapeutic approaches (i.e., checkpoint inhibitors, vaccines, CAR-modified NK or T cells, oncolytic virotherapy) that have been used in an attempt to enhance the immune response against GB, and thereby avoid recidivism or increase survival of GB patients. Finally, we present treatment attempts made using nanotherapies (nanometric structures having active anti-GB agents such as antibodies, chemotherapeutic/anti-angiogenic drugs or sensitizers, radionuclides, and molecules that target GB cellular receptors or open the blood-brain barrier) and non-ionizing energies (laser interstitial thermal therapy, high/low intensity focused ultrasounds, photodynamic/sonodynamic therapies and electroporation). The aim of this review is to discuss the advances and limitations of the current therapies and to present novel approaches that are under development or following clinical trials.
Collapse
Affiliation(s)
- Elena Obrador
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Paz Moreno-Murciano
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
| | - María Oriol-Caballo
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Rafael López-Blanch
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Begoña Pineda
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Julia Lara Gutiérrez-Arroyo
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain; (J.L.G.-A.); (A.L.); (C.M.-C.)
| | - Alba Loras
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain; (J.L.G.-A.); (A.L.); (C.M.-C.)
| | - Luis G. Gonzalez-Bonet
- Department of Neurosurgery, Castellon General University Hospital, 12004 Castellon, Spain;
| | - Conrado Martinez-Cadenas
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain; (J.L.G.-A.); (A.L.); (C.M.-C.)
| | - José M. Estrela
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
- Department of Physiology, Faculty of Pharmacy, University of Valencia, 46100 Burjassot, Spain
| | | |
Collapse
|
20
|
Kudruk S, Forsyth CM, Dion MZ, Hedlund Orbeck JK, Luo J, Klein RS, Kim AH, Heimberger AB, Mirkin CA, Stegh AH, Artzi N. Multimodal neuro-nanotechnology: Challenging the existing paradigm in glioblastoma therapy. Proc Natl Acad Sci U S A 2024; 121:e2306973121. [PMID: 38346200 PMCID: PMC10895370 DOI: 10.1073/pnas.2306973121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2024] Open
Abstract
Integrating multimodal neuro- and nanotechnology-enabled precision immunotherapies with extant systemic immunotherapies may finally provide a significant breakthrough for combatting glioblastoma (GBM). The potency of this approach lies in its ability to train the immune system to efficiently identify and eradicate cancer cells, thereby creating anti-tumor immune memory while minimizing multi-mechanistic immune suppression. A critical aspect of these therapies is the controlled, spatiotemporal delivery of structurally defined nanotherapeutics into the GBM tumor microenvironment (TME). Architectures such as spherical nucleic acids or poly(beta-amino ester)/dendrimer-based nanoparticles have shown promising results in preclinical models due to their multivalency and abilities to activate antigen-presenting cells and prime antigen-specific T cells. These nanostructures also permit systematic variation to optimize their distribution, TME accumulation, cellular uptake, and overall immunostimulatory effects. Delving deeper into the relationships between nanotherapeutic structures and their performance will accelerate nano-drug development and pave the way for the rapid clinical translation of advanced nanomedicines. In addition, the efficacy of nanotechnology-based immunotherapies may be enhanced when integrated with emerging precision surgical techniques, such as laser interstitial thermal therapy, and when combined with systemic immunotherapies, particularly inhibitors of immune-mediated checkpoints and immunosuppressive adenosine signaling. In this perspective, we highlight the potential of emerging treatment modalities, combining advances in biomedical engineering and neurotechnology development with existing immunotherapies to overcome treatment resistance and transform the management of GBM. We conclude with a call to action for researchers to leverage these technologies and accelerate their translation into the clinic.
Collapse
Affiliation(s)
- Sergej Kudruk
- Department of Chemistry, Northwestern University, Evanston, IL60208
- International Institute for Nanotechnology, Northwestern University, Evanston, IL60208
| | - Connor M. Forsyth
- Department of Chemistry, Northwestern University, Evanston, IL60208
- International Institute for Nanotechnology, Northwestern University, Evanston, IL60208
| | - Michelle Z. Dion
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA02115
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Jenny K. Hedlund Orbeck
- Department of Chemistry, Northwestern University, Evanston, IL60208
- International Institute for Nanotechnology, Northwestern University, Evanston, IL60208
| | - Jingqin Luo
- The Brain Tumor Center, Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO63110
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St. Louis, MO63110
| | - Robyn S. Klein
- Department of Medicine, Washington University School of Medicine, St. Louis, MO
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO63110
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO63110
- Center for Neuroimmunology and Neuroinfectious Diseases, Washington University School of Medicine, St. Louis, MO63110
| | - Albert H. Kim
- The Brain Tumor Center, Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO63110
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO63110
| | - Amy B. Heimberger
- Department of Neurological Surgery, Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL60611
| | - Chad A. Mirkin
- Department of Chemistry, Northwestern University, Evanston, IL60208
- International Institute for Nanotechnology, Northwestern University, Evanston, IL60208
| | - Alexander H. Stegh
- The Brain Tumor Center, Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO63110
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO63110
| | - Natalie Artzi
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Medicine, Engineering in Medicine Division, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA02115
| |
Collapse
|
21
|
Liu X, Cheng Y, Mu Y, Zhang Z, Tian D, Liu Y, Hu X, Wen T. Diverse drug delivery systems for the enhancement of cancer immunotherapy: an overview. Front Immunol 2024; 15:1328145. [PMID: 38298192 PMCID: PMC10828056 DOI: 10.3389/fimmu.2024.1328145] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 01/03/2024] [Indexed: 02/02/2024] Open
Abstract
Despite the clear benefits demonstrated by immunotherapy, there is still an inevitable off-target effect resulting in serious adverse immune reactions. In recent years, the research and development of Drug Delivery System (DDS) has received increased prominence. In decades of development, DDS has demonstrated the ability to deliver drugs in a precisely targeted manner to mitigate side effects and has the advantages of flexible control of drug release, improved pharmacokinetics, and drug distribution. Therefore, we consider that combining cancer immunotherapy with DDS can enhance the anti-tumor ability. In this paper, we provide an overview of the latest drug delivery strategies in cancer immunotherapy and briefly introduce the characteristics of DDS based on nano-carriers (liposomes, polymer nano-micelles, mesoporous silica, extracellular vesicles, etc.) and coupling technology (ADCs, PDCs and targeted protein degradation). Our aim is to show readers a variety of drug delivery platforms under different immune mechanisms, and analyze their advantages and limitations, to provide more superior and accurate targeting strategies for cancer immunotherapy.
Collapse
Affiliation(s)
- Xu Liu
- Department of Respiratory and Infectious Disease of Geriatrics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yang Cheng
- Department of Respiratory and Infectious Disease of Geriatrics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yao Mu
- Department of Respiratory and Infectious Disease of Geriatrics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | | | - Dan Tian
- Department of Thoracic Surgery, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yunpeng Liu
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Clinical Cancer Treatment and Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xuejun Hu
- Department of Respiratory and Infectious Disease of Geriatrics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ti Wen
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Clinical Cancer Treatment and Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
22
|
Haskell-Mendoza AP, Srinivasan ES, Gonzalez AT, Reason EH, Jackson JD, Flusche AM, Wachsmuth LP, Lerner E, Underwood D, Buckley ED, Zaidi SE, Herndon JE, Fecci PE. Combination checkpoint blockade and laser interstitial thermal therapy in radiographically progressive non-small cell lung cancer brain metastases. Neurooncol Adv 2024; 6:vdae207. [PMID: 39717437 PMCID: PMC11664258 DOI: 10.1093/noajnl/vdae207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2024] Open
Abstract
Background Laser interstitial thermal therapy (LITT) is a minimally invasive surgical treatment being employed frequently for radiographically progressive brain metastases. Considerable interest exists in combining LITT-mediated in situ vaccination to license immune checkpoint blockade (ICB). No studies have examined the clinical feasibility of this combination in brain metastases. Methods All patients receiving LITT for radiographically progressive non-small cell lung carcinoma (NSCLC) brain metastases at a single center from 2015 to 2023 were retrospectively reviewed. Combination therapy was defined as ICB within 6 weeks of LITT. Clinical data, post-LITT freedom from local progression, and overall survival (OS) were collected. Adverse events (AEs) were evaluated according to Common Terminology Criteria. Results Eighteen patients received LITT + ICB for a total of 19 lesions. The median time between therapies was 2.29 weeks (range 0.85-5.98). In comparison to NSCLC patients receiving LITT alone or with targeted therapy (LITT only) (n = 25), there was no decrement in procedural outcomes. Patients receiving LITT + ICB discontinued steroids at a median of 11 (4-147) days post-LITT vs. 24 (3-242) days for patients receiving LITT only (P = .62). At study cutoff, the local control rate was 18/19 (94.7%) lesions in the LITT + ICB group and 22/25 (88.0%) in the LITT only group. There were 3 and 5 AEs ≥Grade 3 in the LITT + ICB and LITT-only groups, respectively. Conclusions Combination of LITT and ICB does not compromise procedural outcomes or time to steroid discontinuation in NSCLC. Prospective studies are needed to assess biomarkers of immune response.
Collapse
Affiliation(s)
| | - Ethan S Srinivasan
- Department of Neurosurgery, Johns Hopkins University Hospital, Baltimore, Maryland, USA
| | | | - Ellery H Reason
- Duke University School of Medicine, Durham, North Carolina, USA
| | - Joshua D Jackson
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina, USA
| | | | - Lucas P Wachsmuth
- Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Emily Lerner
- Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Delaney Underwood
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Evan D Buckley
- Biostatistics Shared Resource, Duke Cancer Institute, Durham, North Carolina, USA
| | - Saif E Zaidi
- Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina, USA
| | - James E Herndon
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Peter E Fecci
- Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
23
|
Zhou D, Gong Z, Wu D, Ma C, Hou L, Niu X, Xu T. Harnessing immunotherapy for brain metastases: insights into tumor-brain microenvironment interactions and emerging treatment modalities. J Hematol Oncol 2023; 16:121. [PMID: 38104104 PMCID: PMC10725587 DOI: 10.1186/s13045-023-01518-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 12/01/2023] [Indexed: 12/19/2023] Open
Abstract
Brain metastases signify a deleterious milestone in the progression of several advanced cancers, predominantly originating from lung, breast and melanoma malignancies, with a median survival timeframe nearing six months. Existing therapeutic regimens yield suboptimal outcomes; however, burgeoning insights into the tumor microenvironment, particularly the immunosuppressive milieu engendered by tumor-brain interplay, posit immunotherapy as a promising avenue for ameliorating brain metastases. In this review, we meticulously delineate the research advancements concerning the microenvironment of brain metastases, striving to elucidate the panorama of their onset and evolution. We encapsulate three emergent immunotherapeutic strategies, namely immune checkpoint inhibition, chimeric antigen receptor (CAR) T cell transplantation and glial cell-targeted immunoenhancement. We underscore the imperative of aligning immunotherapy development with in-depth understanding of the tumor microenvironment and engendering innovative delivery platforms. Moreover, the integration with established or avant-garde physical methodologies and localized applications warrants consideration in the prevailing therapeutic schema.
Collapse
Affiliation(s)
- Dairan Zhou
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Huangpu District, Shanghai, 200003, People's Republic of China
| | - Zhenyu Gong
- Department of Neurosurgery, Klinikum Rechts Der Isar, Technical University of Munich, Munich, 81675, Germany
| | - Dejun Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, People's Republic of China
| | - Chao Ma
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, People's Republic of China
| | - Lijun Hou
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Huangpu District, Shanghai, 200003, People's Republic of China
| | - Xiaomin Niu
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 241 Huaihai West Road, Xuhui District, Shanghai, 200030, People's Republic of China.
| | - Tao Xu
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Huangpu District, Shanghai, 200003, People's Republic of China.
| |
Collapse
|
24
|
Stamp MEM, Halwes M, Nisbet D, Collins DJ. Breaking barriers: exploring mechanisms behind opening the blood-brain barrier. Fluids Barriers CNS 2023; 20:87. [PMID: 38017530 PMCID: PMC10683235 DOI: 10.1186/s12987-023-00489-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 11/13/2023] [Indexed: 11/30/2023] Open
Abstract
The blood-brain barrier (BBB) is a selectively permeable membrane that separates the bloodstream from the brain. While useful for protecting neural tissue from harmful substances, brain-related diseases are difficult to treat due to this barrier, as it also limits the efficacy of drug delivery. To address this, promising new approaches for enhancing drug delivery are based on disrupting the BBB using physical means, including optical/photothermal therapy, electrical stimulation, and acoustic/mechanical stimulation. These physical mechanisms can temporarily and locally open the BBB, allowing drugs and other substances to enter. Focused ultrasound is particularly promising, with the ability to focus energies to targeted, deep-brain regions. In this review, we examine recent advances in physical approaches for temporary BBB disruption, describing their underlying mechanisms as well as evaluating the utility of these physical approaches with regard to their potential risks and limitations. While these methods have demonstrated efficacy in disrupting the BBB, their safety, comparative efficacy, and practicality for clinical use remain an ongoing topic of research.
Collapse
Affiliation(s)
- Melanie E M Stamp
- Department of Biomedical Engineering, The University of Melbourne, Melbourne, Australia.
- Graeme Clark Institute for Biomedical Engineering, The University of Melbourne, Melbourne, Australia.
| | - Michael Halwes
- Department of Biomedical Engineering, The University of Melbourne, Melbourne, Australia
- Graeme Clark Institute for Biomedical Engineering, The University of Melbourne, Melbourne, Australia
| | - David Nisbet
- Department of Biomedical Engineering, The University of Melbourne, Melbourne, Australia
- Graeme Clark Institute for Biomedical Engineering, The University of Melbourne, Melbourne, Australia
| | - David J Collins
- Department of Biomedical Engineering, The University of Melbourne, Melbourne, Australia
- Graeme Clark Institute for Biomedical Engineering, The University of Melbourne, Melbourne, Australia
| |
Collapse
|
25
|
Chandar JS, Bhatia S, Ingle S, Mendez Valdez MJ, Maric D, Seetharam D, Desgraves JF, Govindarajan V, Daggubati L, Merenzon M, Morell A, Luther E, Saad AG, Komotar RJ, Ivan ME, Shah AH. Laser Interstitial Thermal Therapy Induces Robust Local Immune Response for Newly Diagnosed Glioblastoma With Long-term Survival and Disease Control. J Immunother 2023; 46:351-354. [PMID: 37727953 PMCID: PMC10591996 DOI: 10.1097/cji.0000000000000485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 08/14/2023] [Indexed: 09/21/2023]
Abstract
Laser interstitial thermal therapy (LITT) is a minimally invasive neurosurgical technique used to ablate intra-axial brain tumors. The impact of LITT on the tumor microenvironment is scarcely reported. Nonablative LITT-induced hyperthermia (33-43˚C) increases intra-tumoral mutational burden and neoantigen production, promoting immunogenic cell death. To understand the local immune response post-LITT, we performed longitudinal molecular profiling in a newly diagnosed glioblastoma and conducted a systematic review of anti-tumoral immune responses after LITT. A 51-year-old male presented after a fall with progressive dizziness, ataxia, and worsening headaches with a small, frontal ring-enhancing lesion. After clinical and radiographic progression, the patient underwent stereotactic needle biopsy, confirming an IDH-WT World Health Organization Grade IV Glioblastoma, followed by LITT. The patient was subsequently started on adjuvant temozolomide, and 60 Gy fractionated radiotherapy to the post-LITT tumor volume. After 3 months, surgical debulking was conducted due to perilesional vasogenic edema and cognitive decline, with H&E staining demonstrating perivascular lymphocytic infiltration. Postoperative serial imaging over 3 years showed no evidence of tumor recurrence. The patient is currently alive 9 years after diagnosis. Multiplex immunofluorescence imaging of pre-LITT and post-LITT biopsies showed increased CD8 and activated macrophage infiltration and programmed death ligand 1 expression. This is the first depiction of the in-situ immune response to LITT and the first human clinical presentation of increased CD8 infiltration and programmed death ligand 1 expression in post-LITT tissue. Our findings point to LITT as a treatment approach with the potential for long-term delay of recurrence and improving response to immunotherapy.
Collapse
Affiliation(s)
- Jay S. Chandar
- University of Miami School of Medicine, Department of Neurosurgery, Miami, Florida, United States
- Florida International University Herbert Wertheim College of Medicine, Miami, Florida, United States
| | - Shovan Bhatia
- University of Miami School of Medicine, Department of Neurosurgery, Miami, Florida, United States
| | - Shreya Ingle
- University of Miami School of Medicine, Department of Neurosurgery, Miami, Florida, United States
| | - Mynor J. Mendez Valdez
- University of Miami School of Medicine, Department of Neurosurgery, Miami, Florida, United States
| | - Dragan Maric
- National Institutes of Health, National Institute of Neurological Disorders and Stroke (NINDS), Bethesda, Maryland, United States
| | - Deepa Seetharam
- University of Miami School of Medicine, Department of Neurosurgery, Miami, Florida, United States
| | - Jelisah F. Desgraves
- University of Miami School of Medicine, Department of Neurosurgery, Miami, Florida, United States
| | - Vaidya Govindarajan
- University of Miami School of Medicine, Department of Neurosurgery, Miami, Florida, United States
| | - Lekhaj Daggubati
- University of Miami School of Medicine, Department of Neurosurgery, Miami, Florida, United States
| | - Martin Merenzon
- University of Miami School of Medicine, Department of Neurosurgery, Miami, Florida, United States
| | - Alexis Morell
- University of Miami School of Medicine, Department of Neurosurgery, Miami, Florida, United States
| | - Evan Luther
- University of Miami School of Medicine, Department of Neurosurgery, Miami, Florida, United States
| | - Ali G. Saad
- University of Miami School of Medicine, Department of Neurosurgery, Miami, Florida, United States
| | - Ricardo J. Komotar
- University of Miami School of Medicine, Department of Neurosurgery, Miami, Florida, United States
| | - Michael E. Ivan
- University of Miami School of Medicine, Department of Neurosurgery, Miami, Florida, United States
| | - Ashish H. Shah
- University of Miami School of Medicine, Department of Neurosurgery, Miami, Florida, United States
| |
Collapse
|
26
|
Zhao N, Chung TD, Guo Z, Jamieson JJ, Liang L, Linville RM, Pessell AF, Wang L, Searson PC. The influence of physiological and pathological perturbations on blood-brain barrier function. Front Neurosci 2023; 17:1289894. [PMID: 37937070 PMCID: PMC10626523 DOI: 10.3389/fnins.2023.1289894] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 10/06/2023] [Indexed: 11/09/2023] Open
Abstract
The blood-brain barrier (BBB) is located at the interface between the vascular system and the brain parenchyma, and is responsible for communication with systemic circulation and peripheral tissues. During life, the BBB can be subjected to a wide range of perturbations or stresses that may be endogenous or exogenous, pathological or therapeutic, or intended or unintended. The risk factors for many diseases of the brain are multifactorial and involve perturbations that may occur simultaneously (e.g., two-hit model for Alzheimer's disease) and result in different outcomes. Therefore, it is important to understand the influence of individual perturbations on BBB function in isolation. Here we review the effects of eight perturbations: mechanical forces, temperature, electromagnetic radiation, hypoxia, endogenous factors, exogenous factors, chemical factors, and pathogens. While some perturbations may result in acute or chronic BBB disruption, many are also exploited for diagnostic or therapeutic purposes. The resultant outcome on BBB function depends on the dose (or magnitude) and duration of the perturbation. Homeostasis may be restored by self-repair, for example, via processes such as proliferation of affected cells or angiogenesis to create new vasculature. Transient or sustained BBB dysfunction may result in acute or pathological symptoms, for example, microhemorrhages or hypoperfusion. In more extreme cases, perturbations may lead to cytotoxicity and cell death, for example, through exposure to cytotoxic plaques.
Collapse
Affiliation(s)
- Nan Zhao
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
| | - Tracy D. Chung
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Zhaobin Guo
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
| | - John J. Jamieson
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Lily Liang
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Raleigh M. Linville
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Alex F. Pessell
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Linus Wang
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Peter C. Searson
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
27
|
Wei Z, Yu X, Huang M, Wen L, Lu C. Nanoplatforms Potentiated Ablation-Immune Synergistic Therapy through Improving Local Control and Suppressing Recurrent Metastasis. Pharmaceutics 2023; 15:1456. [PMID: 37242696 PMCID: PMC10224284 DOI: 10.3390/pharmaceutics15051456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/27/2023] [Accepted: 05/07/2023] [Indexed: 05/28/2023] Open
Abstract
Minimally invasive ablation has been widely applied for treatment of various solid tumors, including hepatocellular carcinoma, renal cell carcinoma, breast carcinomas, etc. In addition to removing the primary tumor lesion, ablative techniques are also capable of improving the anti-tumor immune response by inducing immunogenic tumor cell death and modulating the tumor immune microenvironment, which may be of great benefit to inhibit the recurrent metastasis of residual tumor. However, the short-acting activated anti-tumor immunity of post-ablation will rapidly reverse into an immunosuppressive state, and the recurrent metastasis owing to incomplete ablation is closely associated with a dismal prognosis for the patients. In recent years, numerous nanoplatforms have been developed to improve the local ablative effect through enhancing the targeting delivery and combining it with chemotherapy. Particularly, amplifying the anti-tumor immune stimulus signal, modulating the immunosuppressive microenvironment, and improving the anti-tumor immune response with the versatile nanoplatforms have heralded great application prospects for improving the local control and preventing tumor recurrence and distant metastasis. This review discusses recent advances in nanoplatform-potentiated ablation-immune synergistic tumor therapy, focusing on common ablation techniques including radiofrequency, microwave, laser, and high-intensity focused ultrasound ablation, cryoablation, and magnetic hyperthermia ablation, etc. We discuss the advantages and challenges of the corresponding therapies and propose possible directions for future research, which is expected to provide references for improving the traditional ablation efficacy.
Collapse
Affiliation(s)
- Zixuan Wei
- Medical College, Guangxi University, Nanning 530004, China; (Z.W.); (X.Y.)
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Jinan University, Zhuhai 519000, China;
| | - Xiaoya Yu
- Medical College, Guangxi University, Nanning 530004, China; (Z.W.); (X.Y.)
| | - Mao Huang
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Jinan University, Zhuhai 519000, China;
| | - Liewei Wen
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Jinan University, Zhuhai 519000, China;
| | - Cuixia Lu
- Medical College, Guangxi University, Nanning 530004, China; (Z.W.); (X.Y.)
| |
Collapse
|
28
|
Zhu X, Fang Y, Chen Y, Chen Y, Hong W, Wei W, Tu J. Interaction of tumor-associated microglia/macrophages and cancer stem cells in glioma. Life Sci 2023; 320:121558. [PMID: 36889666 DOI: 10.1016/j.lfs.2023.121558] [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: 12/07/2022] [Revised: 02/28/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023]
Abstract
Glioma is the most common tumor of the primary central nervous system, and its malignant phenotype has been shown to be closely related to glioma stem cells (GSCs). Although temozolomide has significantly improved the therapeutic outcome of glioma with a high penetration rate of the blood-brain barrier, resistance is often present in patients. Moreover, evidence has shown that the crosstalk between GSCs and tumor-associated microglia/macrophages (TAMs) affect the clinical occurrence, growth, and multi-tolerance of chemoradiotherapy in gliomas. Here, we highlight its vital roles in the maintenance of the stemness of GSCs and the ability of GSCs to recruit TAMs to the tumor microenvironment and promote their polarization into tumor-promoting macrophages, hence providing groundwork for future research into new treatment strategies of cancer.
Collapse
Affiliation(s)
- Xiangling Zhu
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Yilong Fang
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Yizhao Chen
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Yu Chen
- Department of Gynecology, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Wenming Hong
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wei Wei
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China.
| | - Jiajie Tu
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China.
| |
Collapse
|
29
|
Abstract
Immunotherapy has revolutionized the treatment of patients with cancer. However, promoting antitumour immunity in patients with tumours that are resistant to these therapies remains a challenge. Thermal therapies provide a promising immune-adjuvant strategy for use with immunotherapy, mostly owing to the capacity to reprogramme the tumour microenvironment through induction of immunogenic cell death, which also promotes the recruitment of endogenous immune cells. Thus, thermal immunotherapeutic strategies for various cancers are an area of considerable research interest. In this Review, we describe the role of the various thermal therapies and provide an update on attempts to combine these with immunotherapies in clinical trials. We also provide an overview of the preclinical development of various thermal immuno-nanomedicines, which are capable of combining thermal therapies with various immunotherapy strategies in a single therapeutic platform. Finally, we discuss the challenges associated with the clinical translation of thermal immuno-nanomedicines and emphasize the importance of multidisciplinary and inter-professional collaboration to facilitate the optimal translation of this technology from bench to bedside.
Collapse
|
30
|
Spacca B, Di Maurizio M, Grandoni M, Tempesti S, Genitori L. Laser interstitial thermal therapy (LITT) for pediatric patients affected by intracranial tumors. Front Neurol 2023; 14:1120286. [PMID: 37153686 PMCID: PMC10157164 DOI: 10.3389/fneur.2023.1120286] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 03/16/2023] [Indexed: 05/10/2023] Open
Abstract
Introduction The surgical treatment of brain tumors has evolved over time, offering different strategies tailored to patients and their specific lesions. Among these strategies, Laser Interstitial Thermal Therapy (LITT) is one of the most recent advances in pediatric neurooncological surgery, and its results and evolution are still under assessment. Methods We retrospectively analyzed data from six pediatric patients with deep-seated brain tumors treated with LITT at a single center between November 2019 and June 2022. A total of four patients underwent a stereotaxic biopsy during the same operating session. The indications and preparation for LITT, technical issues, clinical and radiological follow-up, impact on quality of life, and oncological treatment are discussed. Results The mean patient age eight years (ranging from 2 to 11 years). The lesion was thalamic in four patients, thalamo-peduncular in one, and occipital posterior periventricular in one. In total, two patients had been previously diagnosed with low-grade glioma (LGG). Biopsies revealed LGG in two patients, ganglioglioma grade I in one, and diffuse high-grade glioma (HGG) in one. Postoperatively, two patients presented with transient motor deficits. The mean follow-up period was 17 months (ranging from 5 to 32 months). Radiological follow-up showed a progressive reduction of the tumor in patients with LGG. Conclusion Laser interstitial thermal therapy is a promising, minimally invasive treatment for deep-seated tumors in children. The results of lesion reduction appear to be relevant in LGGs and continue over time. It can be used as an alternative treatment for tumors located at sites that are difficult to access surgically or where other standard treatment options have failed.
Collapse
Affiliation(s)
- Barbara Spacca
- Neurosurgery Unit, Meyer Children’s Hospital IRCCS, Florence, Italy
- *Correspondence: Barbara Spacca,
| | | | - Manuela Grandoni
- Neurosurgery Unit, Meyer Children’s Hospital IRCCS, Florence, Italy
| | - Sara Tempesti
- Radiology Unit, Meyer Children’s Hospital IRCCS, Florence, Italy
| | - Lorenzo Genitori
- Neurosurgery Unit, Meyer Children’s Hospital IRCCS, Florence, Italy
| |
Collapse
|
31
|
John C, Jain K, Masanam HB, Narasimhan AK, Natarajan A. Recent Trends and Opportunities for the Targeted Immuno-Nanomaterials for Cancer Theranostics Applications. MICROMACHINES 2022; 13:2217. [PMID: 36557516 PMCID: PMC9781111 DOI: 10.3390/mi13122217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/08/2022] [Accepted: 12/10/2022] [Indexed: 06/17/2023]
Abstract
The targeted delivery of cancer immunotherapies has increased noticeably in recent years. Recent advancements in immunotherapy, particularly in blocking the immune checkpoints (ICs) axis, have shown favorable treatment outcomes for multiple types of cancer including melanoma and non-small-cell lung cancer (NSLC). Engineered micromachines, including microparticles, and nanoplatforms (organic and inorganic), functionalized with immune agonists can effectively deliver immune-targeting molecules to solid tumors. This review focuses on the nanomaterial-based strategies that have shown promise in identifying and targeting various immunological markers in the tumor microenvironment (TME) for cancer diagnosis and therapy. Nanomaterials-based cancer immunotherapy has improved treatment outcomes by triggering an immune response in the TME. Evaluating the expression levels of ICs in the TME also could potentially aid in diagnosing patients who would respond to IC blockade therapy. Detecting immunological checkpoints in the TME using noninvasive imaging systems via tailored nanosensors improves the identification of patient outcomes in immuno-oncology (IO). To enhance patient-specific analysis, lab-on-chip (LOC) technology is a rapid, cost-effective, and accurate way of recapitulating the TME. Such novel nanomaterial-based technologies have been of great interest for testing immunotherapies and assessing biomarkers. Finally, we provide a perspective on the developments in artificial intelligence tools to facilitate ICs-based nano theranostics toward cancer immunotherapy.
Collapse
Affiliation(s)
- Clyde John
- Department of Molecular and Cellular Biology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Kaahini Jain
- Department of Neuroscience, Boston University, Boston, MA 02215, USA
| | - Hema Brindha Masanam
- Advanced Nano-Theranostics (ANTs), Biomaterials Lab, Department of Biomedical Engineering, SRM Institute of Science and Technology, Kattankulathur, Chennai 603203, Tamil Nadu, India
| | - Ashwin Kumar Narasimhan
- Advanced Nano-Theranostics (ANTs), Biomaterials Lab, Department of Biomedical Engineering, SRM Institute of Science and Technology, Kattankulathur, Chennai 603203, Tamil Nadu, India
| | - Arutselvan Natarajan
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology and Bio-X Program, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
32
|
Zhang L, Zhao J, Hu X, Wang C, Jia Y, Zhu C, Xie S, Lee J, Li F, Ling D. A Peritumorally Injected Immunomodulating Adjuvant Elicits Robust and Safe Metalloimmunotherapy against Solid Tumors. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2206915. [PMID: 35986645 DOI: 10.1002/adma.202206915] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Indexed: 06/15/2023]
Abstract
Clinical immunotherapy of solid tumors elicits durable responses only in a minority of patients, largely due to the highly immunosuppressive tumor microenvironment (TME). Although rational combinations of vaccine adjuvants with inflammatory cytokines or immune agonists that relieve immunosuppression represent an appealing therapeutic strategy against solid tumors, there are unavoidable nonspecific toxicities due to the pleiotropy of cytokines and undesired activation of off-target cells. Herein, a Zn2+ doped layered double hydroxide (Zn-LDH) based immunomodulating adjuvant, which not only relieves immunosuppression but also elicits robust antitumor immunity, is reported. Peritumorally injected Zn-LDH sustainably neutralizes acidic TME and releases abundant Zn2+ , promoting a pro-inflammatory network composed of M1-tumor-associated macrophages, cytotoxic T cells, and natural-killer cells. Moreover, the Zn-LDH internalized by tumor cells effectively disrupts endo-/lysosomes to block autophagy and induces mitochondrial damage, and the released Zn2+ activates the cGas-STING signaling pathway to induce immunogenic cell death, which further promotes the release of tumor-associated antigens to induce antigen-specific cytotoxic T lymphocytes. Unprecedentedly, merely injection of Zn-LDH adjuvant, without using any cytotoxic inflammatory cytokines or immune agonists, significantly inhibits the growth, recurrence, and metastasis of solid tumors in mice. This study provides a rational bottom-up design of potent adjuvant for cancer metalloimmunotherapy against solid tumors.
Collapse
Affiliation(s)
- Lingxiao Zhang
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Jing Zhao
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Xi Hu
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, State Key Laboratory of Oncogenes and Related Genes, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
- Department of Clinical Pharmacy, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, P. R. China
| | - Chenhan Wang
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
- Jiangsu Breast Disease Center, the First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, P. R. China
| | - Yingbo Jia
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Chaojie Zhu
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Shangzhi Xie
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Jiyoung Lee
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Fangyuan Li
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
- WLA Laboratories, Shanghai, 201203, P. R. China
| | - Daishun Ling
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, State Key Laboratory of Oncogenes and Related Genes, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
- WLA Laboratories, Shanghai, 201203, P. R. China
| |
Collapse
|
33
|
Emerging translational approaches for brain cancer therapeutics. Adv Drug Deliv Rev 2022; 189:114522. [PMID: 36030017 DOI: 10.1016/j.addr.2022.114522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|