Skip to main content
Top
Published in: Current Treatment Options in Oncology 12/2021

01-12-2021 | Glioblastoma | Neuro-oncology (GJ Lesser, Section Editor)

Role of Laser Interstitial Thermal Therapy in the Management of Primary and Metastatic Brain Tumors

Authors: Kaitlyn Melnick, MD, David Shin, MD, Farhad Dastmalchi, DVM, Zain Kabeer, BA, Maryam Rahman, MD, MS, David Tran, MD, PhD, Ashley Ghiaseddin, MD

Published in: Current Treatment Options in Oncology | Issue 12/2021

Login to get access

Opinion statement

Laser interstitial thermal therapy (LITT) is a minimally invasive treatment option for brain tumors including glioblastoma, other primary central nervous system (CNS) neoplasms, metastases, and radiation necrosis. LITT employs a fiber optic coupled laser delivery probe stabilized via stereotaxis to deliver thermal energy that induces coagulative necrosis in tumors to achieve effective cytoreduction. LITT complements surgical resection, radiation treatment, tumor treating fields, and systemic therapy, especially in patients who are high risk for surgical resection due to tumor location in eloquent regions or poor functional status. These factors must be balanced with the increased rate of cerebral edema post LITT compared to surgical resection. LITT has also been shown to induce transient disruption of the blood–brain barrier (BBB), especially in the peritumoral region, which allows for enhanced CNS delivery of anti-neoplastic agents, thus greatly expanding the armamentarium against brain tumors to include highly effective anti-neoplastic agents that have poor BBB penetration. In addition, hyperthermia-induced immunogenic cell death is another secondary side effect of LITT that opens up immunotherapy as an attractive adjuvant treatment for brain tumors. Numerous large studies have demonstrated the safety and efficacy of LITT against various CNS tumors and as the literature continues to grow on this novel technique so will its indications.
Literature
1.
2.
go back to reference Sugiyama K, Sakai T, Fujishima I, Ryu H, Uemura K, Yokoyama T. Stereotactic interstitial laser-hyperthermia using Nd-YAG laser. Stereotact Funct Neurosurg. 1990;54:501–5.PubMed Sugiyama K, Sakai T, Fujishima I, Ryu H, Uemura K, Yokoyama T. Stereotactic interstitial laser-hyperthermia using Nd-YAG laser. Stereotact Funct Neurosurg. 1990;54:501–5.PubMed
3.
go back to reference Carpentier A, McNichols RJ, Stafford RJ, Itzcovitz J, Guichard J-P, Reizine D, Delaloge S, Vicaut E, Payen D, Gowda A. Real-time magnetic resonance-guided laser thermal therapy for focal metastatic brain tumors. Oper Neurosurg. 2008;63:ONS21–9. Carpentier A, McNichols RJ, Stafford RJ, Itzcovitz J, Guichard J-P, Reizine D, Delaloge S, Vicaut E, Payen D, Gowda A. Real-time magnetic resonance-guided laser thermal therapy for focal metastatic brain tumors. Oper Neurosurg. 2008;63:ONS21–9.
4.
go back to reference Sloan AE, Ahluwalia MS, Valerio-Pascua J, Manjila S, Torchia MG, Jones SE, Sunshine JL, Phillips M, Griswold MA, Clampitt M. Results of the NeuroBlate System first-in-humans Phase I clinical trial for recurrent glioblastoma. J Neurosurg. 2013;118:1202–19.PubMed Sloan AE, Ahluwalia MS, Valerio-Pascua J, Manjila S, Torchia MG, Jones SE, Sunshine JL, Phillips M, Griswold MA, Clampitt M. Results of the NeuroBlate System first-in-humans Phase I clinical trial for recurrent glioblastoma. J Neurosurg. 2013;118:1202–19.PubMed
5.
go back to reference Laurent D, Oliveria SF, Shang M, Bova F, Freedman R, Rahman M. Techniques to ensure accurate targeting for delivery of awake laser interstitial thermotherapy. Oper Neurosurg. 2018;15:454–60. Laurent D, Oliveria SF, Shang M, Bova F, Freedman R, Rahman M. Techniques to ensure accurate targeting for delivery of awake laser interstitial thermotherapy. Oper Neurosurg. 2018;15:454–60.
6.
go back to reference Schwarzmaier H-J, Eickmeyer F, Fiedler VU, Ulrich F. Basic principles of laser induced interstitial thermotherapy in brain tumors. Med Laser Appl. 2002;17:147–58. Schwarzmaier H-J, Eickmeyer F, Fiedler VU, Ulrich F. Basic principles of laser induced interstitial thermotherapy in brain tumors. Med Laser Appl. 2002;17:147–58.
7.
go back to reference Mohammadi AM, Schroeder JL. Laser interstitial thermal therapy in treatment of brain tumors–the NeuroBlate System. Expert Rev Med Devices. 2014;11:109–19.PubMed Mohammadi AM, Schroeder JL. Laser interstitial thermal therapy in treatment of brain tumors–the NeuroBlate System. Expert Rev Med Devices. 2014;11:109–19.PubMed
8.
go back to reference Goldberg SN, Gazelle GS, Mueller PR. Thermal ablation therapy for focal malignancy: a unified approach to underlying principles, techniques, and diagnostic imaging guidance. Am J Roentgenol. 2000;174:323–31. Goldberg SN, Gazelle GS, Mueller PR. Thermal ablation therapy for focal malignancy: a unified approach to underlying principles, techniques, and diagnostic imaging guidance. Am J Roentgenol. 2000;174:323–31.
9.
go back to reference Dewhirst MW, Viglianti B, Lora-Michiels M, Hanson M, Hoopes P. Basic principles of thermal dosimetry and thermal thresholds for tissue damage from hyperthermia. Int J Hyperth. 2003;19:267–94. Dewhirst MW, Viglianti B, Lora-Michiels M, Hanson M, Hoopes P. Basic principles of thermal dosimetry and thermal thresholds for tissue damage from hyperthermia. Int J Hyperth. 2003;19:267–94.
11.
go back to reference Thomsen S. Pathologic analysis of photothermal and photomechanical effects of laser–tissue interactions. Photochem Photobiol. 1991;53:825–35.PubMed Thomsen S. Pathologic analysis of photothermal and photomechanical effects of laser–tissue interactions. Photochem Photobiol. 1991;53:825–35.PubMed
12.
go back to reference Chu KF, Dupuy DE. Thermal ablation of tumours: biological mechanisms and advances in therapy. Nat Rev Cancer. 2014;14:199–208.PubMed Chu KF, Dupuy DE. Thermal ablation of tumours: biological mechanisms and advances in therapy. Nat Rev Cancer. 2014;14:199–208.PubMed
13.
go back to reference Elder JB, Huntoon K, Otero J, Kaya B, Hatef J, Eltobgy M, Lonser RR. Histologic findings associated with laser interstitial thermotherapy for glioblastoma multiforme. Diagn Pathol. 2019;14:1–6. Elder JB, Huntoon K, Otero J, Kaya B, Hatef J, Eltobgy M, Lonser RR. Histologic findings associated with laser interstitial thermotherapy for glioblastoma multiforme. Diagn Pathol. 2019;14:1–6.
14.
go back to reference Leuthardt EC, Duan C, Kim MJ, Campian JL, Kim AH, Miller-Thomas MM, Shimony JS, Tran DD. Hyperthermic laser ablation of recurrent glioblastoma leads to temporary disruption of the peritumoral blood brain barrier. PLoS One. 2016;11:e0148613.PubMedPubMedCentral Leuthardt EC, Duan C, Kim MJ, Campian JL, Kim AH, Miller-Thomas MM, Shimony JS, Tran DD. Hyperthermic laser ablation of recurrent glioblastoma leads to temporary disruption of the peritumoral blood brain barrier. PLoS One. 2016;11:e0148613.PubMedPubMedCentral
15.
go back to reference •Tran D, Campian J, Shimony J, Kim A, Ansstas G, Leuthardt E. Final data analysis of a pilot study testing the efficacy of using laser interstitial thermal therapy (LITT) to induce temporary disruption of the peritumoral blood brain barrier (BBB) to improve effectiveness of BBB-impermeant chemotherapy in recurrent glioblastoma. Neurooncology. 2019;21:vi280. LITT results in disruption of the BBB allowing for chemtherapeutic options not previously feasible. •Tran D, Campian J, Shimony J, Kim A, Ansstas G, Leuthardt E. Final data analysis of a pilot study testing the efficacy of using laser interstitial thermal therapy (LITT) to induce temporary disruption of the peritumoral blood brain barrier (BBB) to improve effectiveness of BBB-impermeant chemotherapy in recurrent glioblastoma. Neurooncology. 2019;21:vi280. LITT results in disruption of the BBB allowing for chemtherapeutic options not previously feasible.
16.
go back to reference Salehi A, Paturu MR, Patel B, Cain MD, Mahlokozera T, Yang AB, Lin T-H, Leuthardt EC, Yano H, Song S-K. Therapeutic enhancement of blood–brain and blood–tumor barriers permeability by laser interstitial thermal therapy. Neurooncol Adv. 2020;2:vdaa071.PubMedPubMedCentral Salehi A, Paturu MR, Patel B, Cain MD, Mahlokozera T, Yang AB, Lin T-H, Leuthardt EC, Yano H, Song S-K. Therapeutic enhancement of blood–brain and blood–tumor barriers permeability by laser interstitial thermal therapy. Neurooncol Adv. 2020;2:vdaa071.PubMedPubMedCentral
17.
go back to reference Slovak R, Ludwig JM, Gettinger SN, Herbst RS, Kim HS. Immuno-thermal ablations–boosting the anticancer immune response. J Immunother Cancer. 2017;5:1–15. Slovak R, Ludwig JM, Gettinger SN, Herbst RS, Kim HS. Immuno-thermal ablations–boosting the anticancer immune response. J Immunother Cancer. 2017;5:1–15.
18.
go back to reference Shin DH, Melnick KF, Tran DD, Ghiaseddin AP. In situ vaccination with laser interstitial thermal therapy augments immunotherapy in malignant gliomas. J Neuro Oncol. 2021;151(1):85–92. Shin DH, Melnick KF, Tran DD, Ghiaseddin AP. In situ vaccination with laser interstitial thermal therapy augments immunotherapy in malignant gliomas. J Neuro Oncol. 2021;151(1):85–92.
19.
go back to reference Srinivasan ES, Sankey EW, Grabowski MM, Chongsathidkiet P, Fecci PE. The intersection between immunotherapy and laser interstitial thermal therapy: a multipronged future of neuro-oncology. Int J Hyperth. 2020;37:27–34. Srinivasan ES, Sankey EW, Grabowski MM, Chongsathidkiet P, Fecci PE. The intersection between immunotherapy and laser interstitial thermal therapy: a multipronged future of neuro-oncology. Int J Hyperth. 2020;37:27–34.
20.
go back to reference Jäättelä M. Heat shock proteins as cellular lifeguards. Ann Med. 1999;31:261–71.PubMed Jäättelä M. Heat shock proteins as cellular lifeguards. Ann Med. 1999;31:261–71.PubMed
21.
go back to reference Ostberg JR, Dayanc BE, Yuan M, Oflazoglu E, Repasky EA. Enhancement of natural killer (NK) cell cytotoxicity by fever-range thermal stress is dependent on NKG2D function and is associated with plasma membrane NKG2D clustering and increased expression of MICA on target cells. J Leukoc Biol. 2007;82:1322–31.PubMed Ostberg JR, Dayanc BE, Yuan M, Oflazoglu E, Repasky EA. Enhancement of natural killer (NK) cell cytotoxicity by fever-range thermal stress is dependent on NKG2D function and is associated with plasma membrane NKG2D clustering and increased expression of MICA on target cells. J Leukoc Biol. 2007;82:1322–31.PubMed
22.
go back to reference Castellino F, Boucher PE, Eichelberg K, Mayhew M, Rothman JE, Houghton AN, Germain RN. Receptor-mediated uptake of antigen/heat shock protein complexes results in major histocompatibility complex class I antigen presentation via two distinct processing pathways. J Exp Med. 2000;191:1957–64.PubMedPubMedCentral Castellino F, Boucher PE, Eichelberg K, Mayhew M, Rothman JE, Houghton AN, Germain RN. Receptor-mediated uptake of antigen/heat shock protein complexes results in major histocompatibility complex class I antigen presentation via two distinct processing pathways. J Exp Med. 2000;191:1957–64.PubMedPubMedCentral
23.
go back to reference Basu S, Binder RJ, Ramalingam T, Srivastava PK. CD91 is a common receptor for heat shock proteins gp96, hsp90, hsp70, and calreticulin. Immunity. 2001;14:303–13.PubMed Basu S, Binder RJ, Ramalingam T, Srivastava PK. CD91 is a common receptor for heat shock proteins gp96, hsp90, hsp70, and calreticulin. Immunity. 2001;14:303–13.PubMed
24.
go back to reference Guo D, Chen Y, Wang S, Yu L, Shen Y, Zhong H, Yang Y. Exosomes from heat-stressed tumour cells inhibit tumour growth by converting regulatory T cells to Th17 cells via IL-6. Immunology. 2018;154:132–43.PubMedPubMedCentral Guo D, Chen Y, Wang S, Yu L, Shen Y, Zhong H, Yang Y. Exosomes from heat-stressed tumour cells inhibit tumour growth by converting regulatory T cells to Th17 cells via IL-6. Immunology. 2018;154:132–43.PubMedPubMedCentral
25.
go back to reference Gastpar R, Gehrmann M, Bausero MA, Asea A, Gross C, Schroeder JA, Multhoff G. Heat shock protein 70 surface-positive tumor exosomes stimulate migratory and cytolytic activity of natural killer cells. Can Res. 2005;65:5238–47. Gastpar R, Gehrmann M, Bausero MA, Asea A, Gross C, Schroeder JA, Multhoff G. Heat shock protein 70 surface-positive tumor exosomes stimulate migratory and cytolytic activity of natural killer cells. Can Res. 2005;65:5238–47.
26.
go back to reference Ivarsson K, Myllymäki L, Jansner K, Stenram U, Tranberg K-G. Resistance to tumour challenge after tumour laser thermotherapy is associated with a cellular immune response. Br J Cancer. 2005;93:435–40.PubMedPubMedCentral Ivarsson K, Myllymäki L, Jansner K, Stenram U, Tranberg K-G. Resistance to tumour challenge after tumour laser thermotherapy is associated with a cellular immune response. Br J Cancer. 2005;93:435–40.PubMedPubMedCentral
27.
go back to reference Isbert C, Ritz JP, Roggan A, Schuppan D, Rühl M, Buhr HJ, Germer CT. Enhancement of the immune response to residual intrahepatic tumor tissue by laser-induced thermotherapy (LITT) compared to hepatic resection. Lasers Surg Med. 2004;35:284–92.PubMed Isbert C, Ritz JP, Roggan A, Schuppan D, Rühl M, Buhr HJ, Germer CT. Enhancement of the immune response to residual intrahepatic tumor tissue by laser-induced thermotherapy (LITT) compared to hepatic resection. Lasers Surg Med. 2004;35:284–92.PubMed
28.
go back to reference Roh JS, Sohn DH. Damage-associated molecular patterns in inflammatory diseases. Immune Netw. 2018;18(4). Roh JS, Sohn DH. Damage-associated molecular patterns in inflammatory diseases. Immune Netw. 2018;18(4).
29.
go back to reference Seong S-Y, Matzinger P. Hydrophobicity: an ancient damage-associated molecular pattern that initiates innate immune responses. Nat Rev Immunol. 2004;4:469–78.PubMed Seong S-Y, Matzinger P. Hydrophobicity: an ancient damage-associated molecular pattern that initiates innate immune responses. Nat Rev Immunol. 2004;4:469–78.PubMed
30.
go back to reference Pålsson-McDermott EM, O’Neill LA. Signal transduction by the lipopolysaccharide receptor, Toll-like receptor-4. Immunology. 2004;113:153–62.PubMedPubMedCentral Pålsson-McDermott EM, O’Neill LA. Signal transduction by the lipopolysaccharide receptor, Toll-like receptor-4. Immunology. 2004;113:153–62.PubMedPubMedCentral
31.
go back to reference Naylor MF, Zhou F, Geister BV, Nordquist RE, Li X, Chen WR. Treatment of advanced melanoma with laser immunotherapy and ipilimumab. J Biophotonics. 2017;10:618–22.PubMed Naylor MF, Zhou F, Geister BV, Nordquist RE, Li X, Chen WR. Treatment of advanced melanoma with laser immunotherapy and ipilimumab. J Biophotonics. 2017;10:618–22.PubMed
32.
go back to reference Liu Y, Chongsathidkiet P, Crawford BM, Odion R, Dechant CA, Kemeny HR, Cui X, Maccarini PF, Lascola CD, Fecci PE. Plasmonic gold nanostar-mediated photothermal immunotherapy for brain tumor ablation and immunologic memory. Immunotherapy. 2019;11:1293–302.PubMedPubMedCentral Liu Y, Chongsathidkiet P, Crawford BM, Odion R, Dechant CA, Kemeny HR, Cui X, Maccarini PF, Lascola CD, Fecci PE. Plasmonic gold nanostar-mediated photothermal immunotherapy for brain tumor ablation and immunologic memory. Immunotherapy. 2019;11:1293–302.PubMedPubMedCentral
33.
go back to reference Overgaard J. The heat is (still) on–the past and future of hyperthermic radiation oncology. Radiother Oncol. 2013;109:185–7.PubMed Overgaard J. The heat is (still) on–the past and future of hyperthermic radiation oncology. Radiother Oncol. 2013;109:185–7.PubMed
34.
go back to reference Kampinga H, Dikomey E. Hyperthermic radiosensitization: mode of action and clinical relevance. Int J Radiat Biol. 2001;77:399–408.PubMed Kampinga H, Dikomey E. Hyperthermic radiosensitization: mode of action and clinical relevance. Int J Radiat Biol. 2001;77:399–408.PubMed
35.
go back to reference Kampinga H, Dynlacht J, Dikomey E. Mechanism of radiosensitization by hyperthermia (43 C) as derived from studies with DNA repair defective mutant cell lines. Int J Hyperth. 2004;20:131–9. Kampinga H, Dynlacht J, Dikomey E. Mechanism of radiosensitization by hyperthermia (43 C) as derived from studies with DNA repair defective mutant cell lines. Int J Hyperth. 2004;20:131–9.
36.
go back to reference Raaphorst GP, Feeley MM. Hyperthermia radiosensitization in human glioma cells comparison of recovery of polymerase activity, survival, and potentially lethal damage repair. Int J Radiat Oncol Biol Phys. 1994;29:133–9.PubMed Raaphorst GP, Feeley MM. Hyperthermia radiosensitization in human glioma cells comparison of recovery of polymerase activity, survival, and potentially lethal damage repair. Int J Radiat Oncol Biol Phys. 1994;29:133–9.PubMed
37.
go back to reference Man J, Shoemake JD, Ma T, Rizzo AE, Godley AR, Wu Q, Mohammadi AM, Bao S, Rich JN, Jennifer SY. Hyperthermia sensitizes glioma stem-like cells to radiation by inhibiting AKT signaling. Can Res. 2015;75:1760–9. Man J, Shoemake JD, Ma T, Rizzo AE, Godley AR, Wu Q, Mohammadi AM, Bao S, Rich JN, Jennifer SY. Hyperthermia sensitizes glioma stem-like cells to radiation by inhibiting AKT signaling. Can Res. 2015;75:1760–9.
38.
go back to reference Raaphorst G, Feeley M, Danjoux C, DaSilva V, Gerig L. Hyperthermia enhancement of radiation response and inhibition of recovery from radiation damage in human glioma cells. Int J Hyperth. 1991;7:629–41. Raaphorst G, Feeley M, Danjoux C, DaSilva V, Gerig L. Hyperthermia enhancement of radiation response and inhibition of recovery from radiation damage in human glioma cells. Int J Hyperth. 1991;7:629–41.
39.
go back to reference Hawasli AH, Bagade S, Shimony JS, Miller-Thomas M, Leuthardt EC. Magnetic resonance imaging-guided focused laser interstitial thermal therapy for intracranial lesions: single-institution series. Neurosurgery. 2013;73:1007–17.PubMed Hawasli AH, Bagade S, Shimony JS, Miller-Thomas M, Leuthardt EC. Magnetic resonance imaging-guided focused laser interstitial thermal therapy for intracranial lesions: single-institution series. Neurosurgery. 2013;73:1007–17.PubMed
40.
go back to reference Bastos DCdA, Rao G, Oliva ICG, Loree JM, Fuentes DT, Stafford RJ, Beechar VB, Weinberg JS, Shah K, Kumar VA. Predictors of local control of brain metastasis treated with laser interstitial thermal therapy. Neurosurgery. 2020;87:112–22.PubMed Bastos DCdA, Rao G, Oliva ICG, Loree JM, Fuentes DT, Stafford RJ, Beechar VB, Weinberg JS, Shah K, Kumar VA. Predictors of local control of brain metastasis treated with laser interstitial thermal therapy. Neurosurgery. 2020;87:112–22.PubMed
41.
go back to reference Mohammadi AM, Hawasli AH, Rodriguez A, Schroeder JL, Laxton AW, Elson P, Tatter SB, Barnett GH, Leuthardt EC. The role of laser interstitial thermal therapy in enhancing progression-free survival of difficult-to-access high-grade gliomas: a multicenter study. Cancer Med. 2014;3:971–9.PubMedPubMedCentral Mohammadi AM, Hawasli AH, Rodriguez A, Schroeder JL, Laxton AW, Elson P, Tatter SB, Barnett GH, Leuthardt EC. The role of laser interstitial thermal therapy in enhancing progression-free survival of difficult-to-access high-grade gliomas: a multicenter study. Cancer Med. 2014;3:971–9.PubMedPubMedCentral
42.
go back to reference Patel P, Patel NV, Danish SF. Intracranial MR-guided laser-induced thermal therapy: single-center experience with the Visualase thermal therapy system. J Neurosurg. 2016;125:853–60.PubMed Patel P, Patel NV, Danish SF. Intracranial MR-guided laser-induced thermal therapy: single-center experience with the Visualase thermal therapy system. J Neurosurg. 2016;125:853–60.PubMed
44.
go back to reference ••Kim AH, Tatter S, Rao G, Prabhu S, Chen C, Fecci P, Chiang V, Smith K, Williams BJ, Mohammadi AM. Laser ablation of abnormal neurological tissue using robotic neuroblate system (LAANTERN): 12-month outcomes and quality of life after brain tumor ablation. Neurosurgery. 2020;87:E338–46. Review of 12 month outcome data of 223 patients treated with LITT at 14 centers. The 1 year survival rate was 71% and was independent of tumor type.PubMedPubMedCentral ••Kim AH, Tatter S, Rao G, Prabhu S, Chen C, Fecci P, Chiang V, Smith K, Williams BJ, Mohammadi AM. Laser ablation of abnormal neurological tissue using robotic neuroblate system (LAANTERN): 12-month outcomes and quality of life after brain tumor ablation. Neurosurgery. 2020;87:E338–46. Review of 12 month outcome data of 223 patients treated with LITT at 14 centers. The 1 year survival rate was 71% and was independent of tumor type.PubMedPubMedCentral
45.
go back to reference ••Shao J, Radakovich NR, Grabowski M, Borghei-Razavi H, Knusel K, Joshi KC, Baha’eddin AM, Hwang L, Barnett GH, Mohammadi AM. Lessons learned in using laser interstitial thermal therapy for treatment of brain tumors: A case series of 238 Patients from a single institution. World Neurosurg. 2020;139:e345–54. Comparison of procedural morbidity and mortality over a ten year period. With time, procedural complications decreased drastically.PubMed ••Shao J, Radakovich NR, Grabowski M, Borghei-Razavi H, Knusel K, Joshi KC, Baha’eddin AM, Hwang L, Barnett GH, Mohammadi AM. Lessons learned in using laser interstitial thermal therapy for treatment of brain tumors: A case series of 238 Patients from a single institution. World Neurosurg. 2020;139:e345–54. Comparison of procedural morbidity and mortality over a ten year period. With time, procedural complications decreased drastically.PubMed
46.
go back to reference Wright J, Chugh J, Wright CH, Alonso F, Hdeib A, Gittleman H, Barnholtz-Sloan J, Sloan AE. Laser interstitial thermal therapy followed by minimal-access transsulcal resection for the treatment of large and difficult to access brain tumors. Neurosurg Focus. 2016;41:E14.PubMed Wright J, Chugh J, Wright CH, Alonso F, Hdeib A, Gittleman H, Barnholtz-Sloan J, Sloan AE. Laser interstitial thermal therapy followed by minimal-access transsulcal resection for the treatment of large and difficult to access brain tumors. Neurosurg Focus. 2016;41:E14.PubMed
47.
go back to reference Pisipati S, Smith KA, Shah K, Ebersole K, Chamoun RB, Camarata PJ. Intracerebral laser interstitial thermal therapy followed by tumor resection to minimize cerebral edema. Neurosurg Focus. 2016;41:E13.PubMed Pisipati S, Smith KA, Shah K, Ebersole K, Chamoun RB, Camarata PJ. Intracerebral laser interstitial thermal therapy followed by tumor resection to minimize cerebral edema. Neurosurg Focus. 2016;41:E13.PubMed
48.
go back to reference Rennert RC, Khan U, Bartek J Jr, Tatter SB, Field M, Toyota B, Fecci PE, Judy K, Mohammadi AM, Landazuri P. Laser ablation of abnormal neurological tissue using robotic neuroblate system (LAANTERN): procedural safety and hospitalization. Neurosurgery. 2020;86:538–47.PubMed Rennert RC, Khan U, Bartek J Jr, Tatter SB, Field M, Toyota B, Fecci PE, Judy K, Mohammadi AM, Landazuri P. Laser ablation of abnormal neurological tissue using robotic neuroblate system (LAANTERN): procedural safety and hospitalization. Neurosurgery. 2020;86:538–47.PubMed
49.
go back to reference Serletis D, Bernstein M. Prospective study of awake craniotomy used routinely and nonselectively for supratentorial tumors. J Neurosurg. 2007;107:1–6.PubMed Serletis D, Bernstein M. Prospective study of awake craniotomy used routinely and nonselectively for supratentorial tumors. J Neurosurg. 2007;107:1–6.PubMed
50.
go back to reference Fadul C, Wood J, Thaler H, Galicich J, Patterson R, Posner J. Morbidity and mortality of craniotomy for excision of supratentorial gliomas. Neurology. 1988;38:1374–1374.PubMed Fadul C, Wood J, Thaler H, Galicich J, Patterson R, Posner J. Morbidity and mortality of craniotomy for excision of supratentorial gliomas. Neurology. 1988;38:1374–1374.PubMed
51.
go back to reference Korinek A, Golmard J, Elcheick A, Bismuth R, Van Effenterre R, Coriat P, Puybasset L. Risk factors for neurosurgical site infections after craniotomy: a critical reappraisal of antibiotic prophylaxis on 4578 patients. Br J Neurosurg. 2005;19:155–62.PubMed Korinek A, Golmard J, Elcheick A, Bismuth R, Van Effenterre R, Coriat P, Puybasset L. Risk factors for neurosurgical site infections after craniotomy: a critical reappraisal of antibiotic prophylaxis on 4578 patients. Br J Neurosurg. 2005;19:155–62.PubMed
52.
go back to reference Korinek A-M. Risk factors for neurosurgical site infections after craniotomy: a prospective multicenter study of 2944 patients. Neurosurgery. 1997;41:1073–81.PubMed Korinek A-M. Risk factors for neurosurgical site infections after craniotomy: a prospective multicenter study of 2944 patients. Neurosurgery. 1997;41:1073–81.PubMed
53.
go back to reference Shi Z-H, Xu M, Wang Y-Z, Luo X-Y, Chen G-Q, Wang X, Wang T, Tang M-Z, Zhou J-X. Post-craniotomy intracranial infection in patients with brain tumors: a retrospective analysis of 5723 consecutive patients. Br J Neurosurg. 2017;31:5–9.PubMed Shi Z-H, Xu M, Wang Y-Z, Luo X-Y, Chen G-Q, Wang X, Wang T, Tang M-Z, Zhou J-X. Post-craniotomy intracranial infection in patients with brain tumors: a retrospective analysis of 5723 consecutive patients. Br J Neurosurg. 2017;31:5–9.PubMed
54.
go back to reference Rahmani R, Tomlinson SB, Santangelo G, Warren KT, Schmidt T, Walter KA, Vates GE. Risk factors associated with early adverse outcomes following craniotomy for malignant glioma in older adults. J Geriatr Oncol. 2020;11:694–700.PubMed Rahmani R, Tomlinson SB, Santangelo G, Warren KT, Schmidt T, Walter KA, Vates GE. Risk factors associated with early adverse outcomes following craniotomy for malignant glioma in older adults. J Geriatr Oncol. 2020;11:694–700.PubMed
55.
go back to reference Cabantog AM, Bernstein M. Complications of first craniotomy for intra-axial brain tumour. Can J Neurol Sci. 1994;21:213–8.PubMed Cabantog AM, Bernstein M. Complications of first craniotomy for intra-axial brain tumour. Can J Neurol Sci. 1994;21:213–8.PubMed
56.
go back to reference Taylor MD, Bernstein M. Awake craniotomy with brain mapping as the routine surgical approach to treating patients with supratentorial intraaxial tumors: a prospective trial of 200 cases. J Neurosurg. 1999;90:35–41.PubMed Taylor MD, Bernstein M. Awake craniotomy with brain mapping as the routine surgical approach to treating patients with supratentorial intraaxial tumors: a prospective trial of 200 cases. J Neurosurg. 1999;90:35–41.PubMed
57.
go back to reference •Leuthardt EC, Voigt J, Kim AH, Sylvester P. A single-center cost analysis of treating primary and metastatic brain cancers with either brain laser interstitial thermal therapy (LITT) or craniotomy. Pharmacoecon Open. 2017;1:53–63. Comparison of cost of LITT versus craniotomy which demonstrated no difference between the two.PubMed •Leuthardt EC, Voigt J, Kim AH, Sylvester P. A single-center cost analysis of treating primary and metastatic brain cancers with either brain laser interstitial thermal therapy (LITT) or craniotomy. Pharmacoecon Open. 2017;1:53–63. Comparison of cost of LITT versus craniotomy which demonstrated no difference between the two.PubMed
58.
go back to reference Voigt JD, Barnett G. The value of using a brain laser interstitial thermal therapy (LITT) system in patients presenting with high grade gliomas where maximal safe resection may not be feasible. Cost Eff Resour Alloc. 2016;14:1–17. Voigt JD, Barnett G. The value of using a brain laser interstitial thermal therapy (LITT) system in patients presenting with high grade gliomas where maximal safe resection may not be feasible. Cost Eff Resour Alloc. 2016;14:1–17.
59.
go back to reference Carpentier A, McNichols RJ, Stafford RJ, Guichard JP, Reizine D, Delaloge S, Vicaut E, Payen D, Gowda A, George B. Laser thermal therapy: Real-time MRI-guided and computer-controlled procedures for metastatic brain tumors. Lasers Surg Med. 2011;43:943–50.PubMed Carpentier A, McNichols RJ, Stafford RJ, Guichard JP, Reizine D, Delaloge S, Vicaut E, Payen D, Gowda A, George B. Laser thermal therapy: Real-time MRI-guided and computer-controlled procedures for metastatic brain tumors. Lasers Surg Med. 2011;43:943–50.PubMed
60.
go back to reference Jethwa PR, Barrese JC, Gowda A, Shetty A, Danish SF. Magnetic resonance thermometry-guided laser-induced thermal therapy for intracranial neoplasms: initial experience. Oper Neurosurg. 2012;71:ons133–45. Jethwa PR, Barrese JC, Gowda A, Shetty A, Danish SF. Magnetic resonance thermometry-guided laser-induced thermal therapy for intracranial neoplasms: initial experience. Oper Neurosurg. 2012;71:ons133–45.
61.
go back to reference Torres-Reveron J, Tomasiewicz HC, Shetty A, Amankulor NM, Chiang VL. Stereotactic laser induced thermotherapy (LITT): a novel treatment for brain lesions regrowing after radiosurgery. J Neurooncol. 2013;113:495–503.PubMed Torres-Reveron J, Tomasiewicz HC, Shetty A, Amankulor NM, Chiang VL. Stereotactic laser induced thermotherapy (LITT): a novel treatment for brain lesions regrowing after radiosurgery. J Neurooncol. 2013;113:495–503.PubMed
62.
go back to reference •Ahluwalia M, Barnett GH, Deng D, Tatter SB, Laxton AW, Mohammadi AM, Leuthardt E, Chamoun R, Judy K, Asher A. Laser ablation after stereotactic radiosurgery: a multicenter prospective study in patients with metastatic brain tumors and radiation necrosis. J Neurosurg. 2018;130:804–11. LAASR study which showed that after SRS, LITT is an effective treatment regardless of if pathology is tumor recurrence or radiation necrosis.PubMed •Ahluwalia M, Barnett GH, Deng D, Tatter SB, Laxton AW, Mohammadi AM, Leuthardt E, Chamoun R, Judy K, Asher A. Laser ablation after stereotactic radiosurgery: a multicenter prospective study in patients with metastatic brain tumors and radiation necrosis. J Neurosurg. 2018;130:804–11. LAASR study which showed that after SRS, LITT is an effective treatment regardless of if pathology is tumor recurrence or radiation necrosis.PubMed
63.
go back to reference Shimony N, Shofty B, Harosh CB, Sitt R, Ram Z, Grossman R. Surgical resection of cerebral metastases leads to faster resolution of peritumoral edema than stereotactic radiosurgery: a volumetric analysis. Ann Surg Oncol. 2017;24:1392–8.PubMed Shimony N, Shofty B, Harosh CB, Sitt R, Ram Z, Grossman R. Surgical resection of cerebral metastases leads to faster resolution of peritumoral edema than stereotactic radiosurgery: a volumetric analysis. Ann Surg Oncol. 2017;24:1392–8.PubMed
64.
go back to reference ••Hong CS, Deng D, Vera A, Chiang VL. Laser-interstitial thermal therapy compared to craniotomy for treatment of radiation necrosis or recurrent tumor in brain metastases failing radiosurgery. J Neurooncol. 2019;142:309–17. There was no major difference in steroid use, neurologic deficits, or survival. This demonstrates that LITT is an equally safe and effective alternative to craniotomy.PubMed ••Hong CS, Deng D, Vera A, Chiang VL. Laser-interstitial thermal therapy compared to craniotomy for treatment of radiation necrosis or recurrent tumor in brain metastases failing radiosurgery. J Neurooncol. 2019;142:309–17. There was no major difference in steroid use, neurologic deficits, or survival. This demonstrates that LITT is an equally safe and effective alternative to craniotomy.PubMed
65.
go back to reference Eichberg DG, Menaker SA, Jermakowicz WJ, Shah AH, Luther EM, Jamshidi AM, Semonche AM, Di L, Komotar RJ, Ivan ME. Multiple iterations of magnetic resonance-guided laser interstitial thermal ablation of brain metastases: single surgeon’s experience and review of the literature. Oper Neurosurg. 2020;19:195–204. Eichberg DG, Menaker SA, Jermakowicz WJ, Shah AH, Luther EM, Jamshidi AM, Semonche AM, Di L, Komotar RJ, Ivan ME. Multiple iterations of magnetic resonance-guided laser interstitial thermal ablation of brain metastases: single surgeon’s experience and review of the literature. Oper Neurosurg. 2020;19:195–204.
66.
go back to reference Swartz LK, Holste KG, Kim MM, Morikawa A, Heth J. Outcomes in patients treated with laser interstitial thermal therapy for primary brain cancer and brain metastases. Oncologist. 2019;24:e1467.PubMedPubMedCentral Swartz LK, Holste KG, Kim MM, Morikawa A, Heth J. Outcomes in patients treated with laser interstitial thermal therapy for primary brain cancer and brain metastases. Oncologist. 2019;24:e1467.PubMedPubMedCentral
67.
go back to reference Sujijantarat N, Hong CS, Owusu KA, Elsamadicy AA, Antonios JP, Koo AB, Baehring JM, Chiang VL. Laser interstitial thermal therapy (LITT) vs. bevacizumab for radiation necrosis in previously irradiated brain metastases. J Neurooncol. 2020;148:641–9.PubMed Sujijantarat N, Hong CS, Owusu KA, Elsamadicy AA, Antonios JP, Koo AB, Baehring JM, Chiang VL. Laser interstitial thermal therapy (LITT) vs. bevacizumab for radiation necrosis in previously irradiated brain metastases. J Neurooncol. 2020;148:641–9.PubMed
68.
go back to reference Jermakowicz WJ, Mahavadi AK, Cajigas I, Dan L, Guerra S, Farooq G, Shah AH, D’Haese PF, Ivan ME, Jagid JR. Predictive modeling of brain tumor laser ablation dynamics. J Neurooncol. 2019;144:193–203.PubMed Jermakowicz WJ, Mahavadi AK, Cajigas I, Dan L, Guerra S, Farooq G, Shah AH, D’Haese PF, Ivan ME, Jagid JR. Predictive modeling of brain tumor laser ablation dynamics. J Neurooncol. 2019;144:193–203.PubMed
69.
go back to reference Hawasli AH, Kim AH, Dunn GP, Tran DD, Leuthardt EC. Stereotactic laser ablation of high-grade gliomas. Neurosurg Focus. 2014;37:E1.PubMed Hawasli AH, Kim AH, Dunn GP, Tran DD, Leuthardt EC. Stereotactic laser ablation of high-grade gliomas. Neurosurg Focus. 2014;37:E1.PubMed
70.
go back to reference Thomas JG, Rao G, Kew Y, Prabhu SS. Laser interstitial thermal therapy for newly diagnosed and recurrent glioblastoma. Neurosurg Focus. 2016;41:E12.PubMed Thomas JG, Rao G, Kew Y, Prabhu SS. Laser interstitial thermal therapy for newly diagnosed and recurrent glioblastoma. Neurosurg Focus. 2016;41:E12.PubMed
71.
go back to reference Mohammadi AM, Sharma M, Beaumont TL, Juarez KO, Kemeny H, Dechant C, Seas A, Sarmey N, Lee BS, Jia X. Upfront magnetic resonance imaging-guided stereotactic laser-ablation in newly diagnosed glioblastoma: a multicenter review of survival outcomes compared to a matched cohort of biopsy-only patients. Neurosurgery. 2019;85:762–72.PubMed Mohammadi AM, Sharma M, Beaumont TL, Juarez KO, Kemeny H, Dechant C, Seas A, Sarmey N, Lee BS, Jia X. Upfront magnetic resonance imaging-guided stereotactic laser-ablation in newly diagnosed glioblastoma: a multicenter review of survival outcomes compared to a matched cohort of biopsy-only patients. Neurosurgery. 2019;85:762–72.PubMed
72.
go back to reference Viozzi I, Guberinic A, Overduin CG, Rovers MM, Ter Laan M. Laser Interstitial Thermal Therapy in Patients with Newly Diagnosed Glioblastoma: A Systematic Review. J Clin Med. 2021;10:355.PubMedPubMedCentral Viozzi I, Guberinic A, Overduin CG, Rovers MM, Ter Laan M. Laser Interstitial Thermal Therapy in Patients with Newly Diagnosed Glioblastoma: A Systematic Review. J Clin Med. 2021;10:355.PubMedPubMedCentral
73.
go back to reference Shah AH, Semonche A, Eichberg DG, Borowy V, Luther E, Sarkiss CA, Morell A, Mahavadi AK, Ivan ME, Komotar RJ. The role of laser interstitial thermal therapy in surgical neuro-oncology: series of 100 consecutive patients. Neurosurgery. 2020;87:266–75.PubMed Shah AH, Semonche A, Eichberg DG, Borowy V, Luther E, Sarkiss CA, Morell A, Mahavadi AK, Ivan ME, Komotar RJ. The role of laser interstitial thermal therapy in surgical neuro-oncology: series of 100 consecutive patients. Neurosurgery. 2020;87:266–75.PubMed
74.
go back to reference Schwarzmaier H-J, Eickmeyer F, von Tempelhoff W, Fiedler VU, Niehoff H, Ulrich SD, Yang Q, Ulrich F. MR-guided laser-induced interstitial thermotherapy of recurrent glioblastoma multiforme: preliminary results in 16 patients. Eur J Radiol. 2006;59:208–15.PubMed Schwarzmaier H-J, Eickmeyer F, von Tempelhoff W, Fiedler VU, Niehoff H, Ulrich SD, Yang Q, Ulrich F. MR-guided laser-induced interstitial thermotherapy of recurrent glioblastoma multiforme: preliminary results in 16 patients. Eur J Radiol. 2006;59:208–15.PubMed
75.
go back to reference Sharma M, Schroeder JL, Elson P, Meola A, Barnett GH, Vogelbaum MA, Suh JH, Chao ST, Mohammadi AM, Stevens GH. Outcomes and prognostic stratification of patients with recurrent glioblastoma treated with salvage stereotactic radiosurgery. J Neurosurg. 2018;131:489–99.PubMed Sharma M, Schroeder JL, Elson P, Meola A, Barnett GH, Vogelbaum MA, Suh JH, Chao ST, Mohammadi AM, Stevens GH. Outcomes and prognostic stratification of patients with recurrent glioblastoma treated with salvage stereotactic radiosurgery. J Neurosurg. 2018;131:489–99.PubMed
76.
go back to reference Kamath AA, Friedman DD, Akbari SHA, Kim AH, Tao Y, Luo J, Leuthardt EC. Glioblastoma treated with magnetic resonance imaging-guided laser interstitial thermal therapy: safety, efficacy, and outcomes. Neurosurgery. 2019;84:836–43.PubMed Kamath AA, Friedman DD, Akbari SHA, Kim AH, Tao Y, Luo J, Leuthardt EC. Glioblastoma treated with magnetic resonance imaging-guided laser interstitial thermal therapy: safety, efficacy, and outcomes. Neurosurgery. 2019;84:836–43.PubMed
77.
go back to reference ••Montemurro N, Anania Y, Cagnazzo F, Perrini P. Survival outcomes in patients with recurrent glioblastoma treated with Laser Interstitial Thermal Therapy (LITT): A systematic review. Clin Neurol Neurosurg. 2020;195:105942. Review of 17 studies on 203 patients with recurrent glioblastoma. The overall survival after LITT for these patients was 10.2 months.PubMed ••Montemurro N, Anania Y, Cagnazzo F, Perrini P. Survival outcomes in patients with recurrent glioblastoma treated with Laser Interstitial Thermal Therapy (LITT): A systematic review. Clin Neurol Neurosurg. 2020;195:105942. Review of 17 studies on 203 patients with recurrent glioblastoma. The overall survival after LITT for these patients was 10.2 months.PubMed
78.
go back to reference Ruben JD, Dally M, Bailey M, Smith R, McLean CA, Fedele P. Cerebral radiation necrosis: incidence, outcomes, and risk factors with emphasis on radiation parameters and chemotherapy. Int J Radiat Oncol Biol Phys. 2006;65:499–508.PubMed Ruben JD, Dally M, Bailey M, Smith R, McLean CA, Fedele P. Cerebral radiation necrosis: incidence, outcomes, and risk factors with emphasis on radiation parameters and chemotherapy. Int J Radiat Oncol Biol Phys. 2006;65:499–508.PubMed
79.
go back to reference Levin VA, Bidaut L, Hou P, Kumar AJ, Wefel JS, Bekele BN, Prabhu S, Loghin M, Gilbert MR, Jackson EF. Randomized double-blind placebo-controlled trial of bevacizumab therapy for radiation necrosis of the central nervous system. Int J Radiat Oncol Biol Phys. 2011;79:1487–95.PubMedPubMedCentral Levin VA, Bidaut L, Hou P, Kumar AJ, Wefel JS, Bekele BN, Prabhu S, Loghin M, Gilbert MR, Jackson EF. Randomized double-blind placebo-controlled trial of bevacizumab therapy for radiation necrosis of the central nervous system. Int J Radiat Oncol Biol Phys. 2011;79:1487–95.PubMedPubMedCentral
80.
go back to reference Rao MS, Hargreaves EL, Khan AJ, Haffty BG, Danish SF. Magnetic resonance-guided laser ablation improves local control for postradiosurgery recurrence and/or radiation necrosis. Neurosurgery. 2014;74:658–67.PubMed Rao MS, Hargreaves EL, Khan AJ, Haffty BG, Danish SF. Magnetic resonance-guided laser ablation improves local control for postradiosurgery recurrence and/or radiation necrosis. Neurosurgery. 2014;74:658–67.PubMed
81.
go back to reference Rahmathulla G, Recinos PF, Valerio JE, Chao S, Barnett GH. Laser interstitial thermal therapy for focal cerebral radiation necrosis: a case report and literature review. Stereotact Funct Neurosurg. 2012;90:192–200.PubMed Rahmathulla G, Recinos PF, Valerio JE, Chao S, Barnett GH. Laser interstitial thermal therapy for focal cerebral radiation necrosis: a case report and literature review. Stereotact Funct Neurosurg. 2012;90:192–200.PubMed
82.
go back to reference Rammo R, Asmaro K, Schultz L, Scarpace L, Siddiqui S, Walbert T, Kalkanis S, Lee I. The safety of magnetic resonance imaging-guided laser interstitial thermal therapy for cerebral radiation necrosis. J Neurooncol. 2018;138:609–17.PubMed Rammo R, Asmaro K, Schultz L, Scarpace L, Siddiqui S, Walbert T, Kalkanis S, Lee I. The safety of magnetic resonance imaging-guided laser interstitial thermal therapy for cerebral radiation necrosis. J Neurooncol. 2018;138:609–17.PubMed
83.
go back to reference Ivan ME, Diaz RJ, Berger MH, Basil GW, Osiason DA, Plate T, Wallo A, Komotar RJ. Magnetic resonance–guided laser ablation for the treatment of recurrent dural-based lesions: a series of five cases. World Neurosurg. 2017;98:162–70.PubMed Ivan ME, Diaz RJ, Berger MH, Basil GW, Osiason DA, Plate T, Wallo A, Komotar RJ. Magnetic resonance–guided laser ablation for the treatment of recurrent dural-based lesions: a series of five cases. World Neurosurg. 2017;98:162–70.PubMed
Metadata
Title
Role of Laser Interstitial Thermal Therapy in the Management of Primary and Metastatic Brain Tumors
Authors
Kaitlyn Melnick, MD
David Shin, MD
Farhad Dastmalchi, DVM
Zain Kabeer, BA
Maryam Rahman, MD, MS
David Tran, MD, PhD
Ashley Ghiaseddin, MD
Publication date
01-12-2021
Publisher
Springer US
Published in
Current Treatment Options in Oncology / Issue 12/2021
Print ISSN: 1527-2729
Electronic ISSN: 1534-6277
DOI
https://doi.org/10.1007/s11864-021-00912-6

Other articles of this Issue 12/2021

Current Treatment Options in Oncology 12/2021 Go to the issue

Gynecologic Cancers (LA Cantrell, Section Editor)

Options for the Treatment of Mucinous Ovarian Carcinoma

Palliative and Supportive Care (MP Davis, Section Editor)

Treating Chronic Pain with Buprenorphine—The Practical Guide

Neuro-oncology (GJ Lesser, Section Editor)

Systemic Therapy for Lung Cancer Brain Metastases

Head and Neck Cancer (JL Geiger, Section Editor)

Overview and Emerging Trends in the Treatment of Osteoradionecrosis

Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine