American Journal of Medicine and Medical Sciences

p-ISSN: 2165-901X    e-ISSN: 2165-9036

2024;  14(4): 874-878

doi:10.5923/j.ajmms.20241404.16

Received: Mar. 2, 2024; Accepted: Mar. 26, 2024; Published: Apr. 3, 2024

 

Chemotherapy Delivered Through Intra-Arterial Route for Brain Gliomas: An Overview of Current Strategies and Future Prospects

B. F. Mahmudov1, U. U. Altybaev1, G. N. Saidov2, S. S. Akhmedov1

1Republican Specialized Scientific and Practical Medical Center of Neurosurgery, Tashkent, Uzbekistan

2Bukhara Branch of the Republican Specialized Scientific and Practical Medical Center of Oncology and Radiology, Bukhara, Uzbekistan

Copyright © 2024 The Author(s). Published by Scientific & Academic Publishing.

This work is licensed under the Creative Commons Attribution International License (CC BY).
http://creativecommons.org/licenses/by/4.0/

Abstract

High-grade glial tumors pose formidable challenges in oncology, characterized by infiltrative growth, proliferative activity, and a propensity for relapse. The standard Stupp protocol, combining radiotherapy with temozolomide, remains suboptimal, with recurrence rates underscoring the critical need for novel therapeutic strategies. Intra-arterial chemotherapy delivery emerges as a promising avenue, offering the potential to achieve elevated local drug concentrations at tumor sites while mitigating systemic side effects. This comprehensive review navigates the historical context of intra-arterial therapy, emphasizing its evolution since the pioneering experiments of Bierman and Klopp in the 1950s. Contemporary advancements, facilitated by radioimaging and endovascular instruments, showcase renewed interest, with approximately 3,000 global procedures annually. However, the blood-brain barrier persists as a formidable hurdle, necessitating exploration of barrier disruption techniques. Insights from phase I studies affirm the safety and tolerability of intra-arterial chemotherapy, prompting the imperative for phase 3 validations. Future research trajectories spotlight the optimization of therapeutic combinations, integration of advanced imaging for precise vascular target identification, and the development of quantitative monitoring methods for direct drug delivery assessment. This article significantly contributes to intra-arterial therapy knowledge, acting as a linchpin for innovative treatment strategies. The potential to enhance prognoses in high-grade glial tumors underscores the relevance of this review, offering a foundation for continued advancements in glioma treatment modalities.

Keywords: Intra-arterial chemotherapy, Glial tumors, Glioblastoma, Anaplastic astrocytoma

Cite this paper: B. F. Mahmudov, U. U. Altybaev, G. N. Saidov, S. S. Akhmedov, Chemotherapy Delivered Through Intra-Arterial Route for Brain Gliomas: An Overview of Current Strategies and Future Prospects, American Journal of Medicine and Medical Sciences, Vol. 14 No. 4, 2024, pp. 874-878. doi: 10.5923/j.ajmms.20241404.16.

1. Introduction

Intracerebral glial tumors arising from neuroglial cells pose a formidable challenge in contemporary oncology due to their infiltrative growth, high proliferative activity, and propensity for relapse. The incidence of primary central nervous system (CNS) tumors exhibits regional variability, ranging from 5 to 14 cases per 100 thousand population. In 2014, Russia reported an incidence of 5.55 cases per 100 thousand population, while developed countries recorded figures between 8.4 to 11.8 in men and 5.8 to 9.3 in women [4,7,13,18].
Since the introduction of the Stupp protocol in 2005, incorporating radiotherapy with oral temozolomide, this approach has become the standard for treating glial brain tumors [6,7,9]. Despite these efforts, a majority of patients experience tumor recurrence within six months after treatment, underscoring the need for novel therapeutic approaches. Existing methods, unfortunately, offer limited progress, rendering glial tumors largely incurable.
Traditional chemotherapy faces challenges such as restricted penetration of the blood-brain barrier and elevated systemic toxicity, diminishing its efficacy [1,19,44]. Consequently, the scientific community explores alternative drug delivery methods, including intra-arterial delivery. Techniques like intrathecal, intracavitary, and convection-enhanced delivery are under investigation, offering potential in glial tumor treatment [2,3,7].
Intra-arterial chemotherapy, proposed in the 1950s, involves directly delivering drugs to the tumor through arterial vessels [15,16,18,22,38]. Initial limitations stemmed from the neurotoxicity of available drugs, hindering research and development.
Advancements in radioimaging and endovascular instruments have enhanced the accuracy and safety of intra-arterial drug delivery. Approximately 3,000 such procedures are now conducted globally, reflecting renewed interest. Improved technologies enable precise catheter guidance and cerebral blood flow modification, opening new avenues for glial tumor treatment [44,45].
However, the blood-brain barrier remains a significant hurdle for direct drug delivery to brain tissue. Techniques like mannitol-induced barrier disruption require further evaluation for efficacy and safety.
Exploring various approaches, intra-arterial chemotherapy for glial tumors considers not only traditional drugs but also antibodies, engineered cells, viruses, and radiotherapies. These innovative strategies hold promise, but their clinical implementation demands thorough investigation.
A review of research in this field contextualizes the historical development of intra-arterial therapy for glial tumors, delineates past challenges faced by researchers, and outlines future prospects for this treatment modality. Emphasizing the need for ongoing research to enhance delivery methods, mitigate toxicity, and improve overall effectiveness in glial tumor treatment, this review underscores the evolving landscape of intra-arterial chemotherapy [11,14,16,18].
The inception of intra-arterial chemotherapy in the 1950s saw pioneering experiments by Bierman, Klopp, and colleagues, exploring the direct delivery of high doses of nitrogen mustard to liver tumors through arterial blood supply. Despite initial promise in rabbit experiments, the use of nitrogen mustard proved ineffective due to low therapeutic benefits and severe hematopoietic system effects. In the 1970s, Ekman demonstrated the potential of intra-arterial chemotherapy for achieving higher drug concentrations in targeted tumors, while Stanley Rapaport investigated blood-brain barrier permeability mechanisms, emphasizing the role of tight junctions [12,13,15,19].
A pivotal study by Levine and collaborators in 1978 revealed significantly enhanced drug delivery to target sites with intra-arterial administration compared to intravenous delivery, affirming the efficacy of this method for chemotherapy delivery to blood vessels.

2. Intra-arterial Drug Infusion: Overcoming Blood-Brain Barrier Challenges

The blood-brain barrier (BBB) stands as a formidable shield, safeguarding the brain from toxins and pathogens, yet concurrently creating formidable barriers for drug delivery, notably chemotherapy drugs, to brain tumors. This selective barrier effectively hinders the passage of molecules with a molecular weight exceeding 180 Da, restricting the access of many chemotherapeutic agents within the range of 200-1200 Da. The challenge of traversing the BBB is particularly pronounced in glioblastoma and other brain tumors, necessitating the direct delivery of drugs to the disease site at concentrations conducive to effective treatment [24,28,29].
In response to these challenges, diverse methods have emerged to enhance drug delivery across the BBB. One such method involves intra-arterial infusion of a hyperosmotic solution, such as mannitol, which transiently opens the tight junctions between endothelial cells, augmenting barrier permeability. While this approach holds promise in improving the delivery of chemotherapeutic agents to brain tumors, its implementation demands meticulous monitoring to avert potential neurological complications.
Distinct characteristics of the BBB and the brain-blood-tumor barrier (BTB) underscore the imperative for innovative approaches in the treatment of malignant gliomas. Elevated expression of vascular endothelial growth factor (VEGF) and angiogenesis contribute to the formation of aberrant vessels in tumors, intensifying the complexities of drug delivery. While high-grade gliomas may exhibit partial permeability in the barrier, low-grade gliomas present a nearly fully functional BBB, heightening the challenge of chemotherapy delivery [25,27,30].
Continual research in drug delivery methodologies across the BBB and BCOM [brain-blood-tumor barrier] includes the exploration of microcatheters, nanotechnology, and other innovative strategies. These evolving techniques hold the promise of providing novel avenues for treating brain tumors by enhancing drug penetration through these barriers, ultimately amplifying therapeutic efficacy [31,32].

3. Intra-arterial Chemoinfusion for Brain Tumors: Unlocking Therapeutic Potential

Intra-arterial chemoinfusion (IAC) emerges as a highly promising avenue for treating brain tumors, notably glioblastoma multiforme, owing to its capability to directly deliver elevated drug concentrations to the tumor site while mitigating systemic side effects. This holds particular significance for glioblastoma multiforme, renowned for its aggressiveness and resistance to conventional treatments like surgery, radiotherapy, and systemic chemotherapy, often hindered by the formidable blood-brain barrier (BBB) [33,34].
Mannitol serves as an agent to disrupt the BBB, heightening its permeability and facilitating more effective delivery of chemotherapy drugs to tumor tissue. Clinical trials exploring IAC chemoinfusion with diverse chemotherapeutic agents, including platinum analogs, methotrexate, vincristine, and novel antibodies like bevacizumab and cetuximab, underscore the potential of this approach in glioblastoma multiforme treatment [32,35,36].
Monoclonal antibodies such as bevacizumab, blocking VEGF action, and cetuximab, inhibiting EGFR, demonstrate promising results in combination therapy for brain tumors. Particularly, when combined with other treatments, these agents show potential to enhance the prognosis for glioblastoma multiforme patients [32,35].
While intra-arterial chemoinfusion presents new avenues for enhancing survival and quality of life in brain tumor patients, further research is imperative to optimize treatment regimens, identify optimal drug combinations, and minimize the risk of side effects.

4. Mechanisms of Action for Bevacizumab and Cetuximab

Bevacizumab disrupts VEGF-A binding to endothelial cell receptors, suppressing angiogenesis pivotal for tumor growth and spread. Its ability to normalize tumor vasculature enhances oxygen delivery, reduces permeability, and amplifies the efficacy of complementary therapies like chemotherapy and radiotherapy. Studies validate its impact on improving progression-free survival in diverse cancers, including glioblastoma multiforme [34,36,38].
Cetuximab inhibits epidermal growth factor receptor (EGFR), often overexpressed in tumor cells, curtailing signaling pathways crucial for tumor growth, angiogenesis, and metastasis. Effective in certain cancers, especially KRAS wild-type colorectal cancer, cetuximab is under investigation for glioblastoma multiforme [39,41].

5. Synergistic Potential of Bevacizumab and Cetuximab

The combined use of bevacizumab and cetuximab may yield a synergistic effect, concurrently suppressing angiogenesis and tumor growth by targeting distinct molecular pathways. This approach holds promise for improving outcomes in tumors resistant to standard therapies [24,42].

6. Consideration of Side Effects

Despite potential benefits, careful consideration of side effects is crucial. Bevacizumab may contribute to hypertension, heightened bleeding risk, and thromboembolic events, while cetuximab is associated with skin reactions and infections. Inclusion of these drugs in treatment regimens necessitates a meticulous benefit-risk analysis tailored to each patient [43,45].

7. Conclusions

Intra-arterial chemotherapy delivery emerges as a promising methodology in the treatment arsenal for high-grade glial tumors, facilitating the attainment of concentrated drug levels at the tumor site while curbing systemic side effects. While phase I studies underscore the safety and favorable tolerability of this approach, the imperative confirmation of its efficacy demands robust phase 3 investigations.
The authors adeptly chart key trajectories for future research, with a focus on optimizing therapeutic amalgamations, leveraging advanced imaging techniques for precise identification of tumor vascular supply target areas, and formulating quantitative methods for monitoring drug delivery directly to tumor tissues.
This article stands as a significant contribution to the knowledge repository concerning intra-arterial therapy for glial tumors. It holds the potential to serve as a linchpin for the development of innovative treatment strategies poised to elevate the prognosis for patients grappling with these formidable diseases.

References

[1]  Lesniak WG, Chu C, Jablonska A, Du Y, Pomper MG, Walczak P, et al. A distinct advantage to intraarterial delivery of ^89Zr-bevacizumab in PET imaging of mice with and without osmotic opening of the blood-brain barrier. J Nucl Med. 2019; 60(5): 617–22. doi: 10.2967/jnumed.118.218792.
[2]  Baumgarten L, Brucker D, Tirniceru A, Kienast Y, Grau S, Burgold S, et al. Bevacizumab has differential and dose-dependent effects on glioma blood vessels and tumor cells. Clin Cancer Res. 2011; 17(19): 6192–205. doi: 10.1158/1078-0432.CCR-10-1868.
[3]  Reifenberger G, Wirsching HG, Knobbe-Thomsen CB, Weller M. Advances in the molecular genetics of gliomas - implications for classification and therapy. Nat Rev Clin Oncol. 2017; 14(7): 434–52. doi: 10.1038/nrclinonc.2016.204.
[4]  Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries [published correction appears in CA Cancer J Clin. CA Cancer J Clin. 2020;68(6):394–424. doi: 10.3322/caac.21492.
[5]  Jacques TS, Swales A, Brzozowski MJ, Henriquez NV, Linehan JM, Mirzadeh Z, et al. Combinations of genetic mutations in the adult neural stem cell compartment determine brain tumor phenotypes. EMBO J. 2010; 29: 222–35. doi: 10.1038/emboj.2009.327.
[6]  Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005; 352(10): 987–96. doi: 10.1056/NEJMoa043330.
[7]  Kopecka J, Riganti C. Overcoming drug resistance in glioblastoma: new options in sight? Cancer Drug Resist. 2021; 4: 512–6. doi: 10.20517/cdr.2021.03.
[8]  Fakhoury M. Drug delivery approaches for the treatment of glioblastoma multiforme. Artif Cells Nanomed Biotechnol. 2016; 44(6): 1365–73. doi: 10.3109/21691401.2015.1052467.
[9]  Chen W, Wu Q, Mo L, Nassi M. Intra-arterial chemotherapy is not superior to intravenous chemotherapy for malignant gliomas: a systematic review and meta-analysis. Eur Neurol. 2013; 70(1-2): 124–32. doi: 10.1159/000346580.
[10]  Burkhardt JK, Riina HA, Shin BJ, Moliterno JA, Hofstetter CP, Boockvar JA. Intra-arterial chemotherapy for malignant gliomas: a critical analysis [published correction appears in Interv Neuroradiol. Interv Neuroradiol. 2011; 17(3): 286–95: 506. doi: 10.1177/159101991101700302.
[11]  D'Amico RS, Khatri D, Reichman N, Patel NV, Wong T, Fraling SH, et al. Super selective intra-arterial cerebral infusion of modern chemotherapeutics after blood-brain barrier disruption: where are we now, and where we are going [published correction appears in J Neurooncol. J Neurooncol. 2020; 147(2): 261–78. doi: 10.1007/s11060-020-03435-6.
[12]  Basso U, Lonardi S, Brandes AA. Is intra-arterial chemotherapy useful in high-grade gliomas? Expert Rev Anticancer Ther. 2002; 2(5): 507–19. doi: 10.1586/14737140.2.5.507.
[13]  Bierman HR, Byron RL Jr, Miller ER, Shimkin MB. Effects of intra-arterial administration of nitrogen mustard. Am J Med. 1950; 8: 535. doi: 10.1016/0002-9343(50)90263-4.
[14]  Bierman HR, Byron RL Jr, Kelly KH. Therapy of inoperable visceral and regional metastases by intra-arterial catheterization in man. Cancer Res. 1951; 11: 236.
[15]  Klopp C.T., Alford T.C., Bateman J, Berry G.N., Winship T. Fractionated intra-arterial cancer chemotherapy with methyl bis amine hydrochloride; A preliminary report. Ann Surg. 1950; 132: 811–32. doi: 10.1097/00000658-195010000-00018.
[16]  French JD, West PM, Von Amerongen FK, Magoun HW. Effects of intracarotid administration of nitrogen mustard on normal brain and brain tumors. J Neurosurg. 1952;9:378–89. doi: 10.3171/jns.1952.9.4.0378.
[17]  Wilson CB. Chemotherapy of brain tumors by continuous arterial infusion. Surgery. 1964; 55: 640–65321.
[18]  Owens G, Javid R, Belmusto L, Bender M, Blau M. Intra-arterial vincristine therapy of primary gliomas. 1965; 18: 756–60. doi: 10.1002/1097-0142(196506) 18:63.0.CO;2-#.
[19]  Eckman WW, Patlak CS, Fenstermacher JD. A critical evaluation of the principles governing the advantages of intra-arterial infusions. J Pharmacokinet Biopharm. 1974; 2: 257–85. doi: 10.1007/BF01059765.
[20]  Rapoport SI, Hori M, Klatzo I. Testing of a hypothesis for osmotic opening of the blood-brain barrier. Am J Physiol. 1972; 223: 323–31. doi: 10.1152/ajplegacy.1972.223.2.323.
[21]  Neuwelt EA, Maravilla KR, Frenkel EP, Rapaport SI, Hill SA, Barnett PA. Osmotic blood-brain barrier disruption: Computerized tomographic monitoring of chemotherapeutic agent delivery. J Clin Invest. 1979; 64(2): 684–8. doi: 10.1172/JCI109509.
[22]  Neuwelt EA, Frenkel EP, Diehl JT, Maravilla KR, Vu LH, Clark WK, et al. Osmotic blood-brain barrier disruption: A new means of increasing chemotherapeutic agent delivery. Trans Am Neurol Assoc. 1979; 104: 256–60.
[23]  Neuwelt EA, Glasberg M, Diehl J, Frenkel EP, Barnett P. Osmotic blood-brain barrier disruption in the posterior fossa of the dog. J Neurosurg. 1981; 55: 742–8. doi: 10.3171/jns.1981.55.5.0742.
[24]  Neuwelt EA, Frenkel EP, D’Agostino AN, Carney DN, Minna JD, Barnett PA, et al. Growth of human lung tumor in the brain of the nude rat as a model to evaluate antitumor agent delivery across the blood-brain barrier. Cancer Res. 1985; 45: 2827–33.
[25]  Neuwelt EA, Barnett PA, McCormick CI, Remsen LG, Kroll RA, Sexton G. Differential permeability of a human brain tumor xenograft in the nude rat: impact of tumor size and method of administration on optimizing delivery of biologically diverse agents. Clin Cancer Res. 1998; 4: 1549–55.
[26]  Levin VA, Kabra PM, Freeman-Dove MA. Pharmacokinetics of intracarotid artery ^14C-BCNU in the squirrel monkey. J Neurosurg. 1978; 48(4): 587–93. doi: 10.3171/jns.1978.48.4.0587.
[27]  Greenberg HS, Ensminger WD, Chandler WF, Layton PB, Junck L, Knake J, et al. Intra-arterial BCNU chemotherapy for treatment of malignant gliomas of the central nervous system. J Neurosurg. 1984; 61: 423–9. doi: 10.3171/jns.1984.61.3.0423.
[28]  Fenstermacher JD, Johnson JA. Filtration and reflection coefficients of the rabbit blood-brain barrier. Am J Physiol. 1966; 211: 341–6. doi: 10.1152/ajplegacy.1966.211.2.341.
[29]  Tellingen O, Yetkin-Arik B, de Gooijer MC, Wesseling P, Wurdinger T, de Vries HE. Overcoming the blood-brain tumor barrier for effective glioblastoma treatment. Drug Resist Updat. 2015; 19: 1–12. doi: 10.1016/j.drup.2015.02.002.
[30]  Ningaraj NS, Rao M, Hashizume K, Asotra K, Black KL. Regulation of blood-brain tumor barrier permeability by calcium-activated potassium channels. J Pharmacol Exp Ther. 2002; 301(3): 838–51. doi: 10.1124/jpet.301.3.838.
[31]  Siegal T, Rubinstein R, Bokstein F, Schwartz A, Lossos A, Shalom E, et al. In vivo assessment of the window of barrier opening after osmotic blood-brain barrier disruption in humans. J Neurosurg. 2000; 92(4): 599–605. doi: 10.3171/jns.2000.92.4.0599.
[32]  Fortin D, Desjardins A, Benko A, Niyonsega T, Boudrias M. Enhanced chemotherapy delivery by intraarterial infusion and blood-brain barrier disruption in malignant brain tumors: the Sherbrooke experience. Cancer. 2005; 103: 2606–2. doi: 10.1002/cncr.21112.
[33]  Nakagawa H, Groothuis D, Blasberg RG. The effect of graded hypertonic intracarotid infusions on drug delivery to experimental RG-2 gliomas. Neurology. 1984; 34: 1571–81. doi: 10.1212/wnl.34.12.1571.
[34]  Groothuis DR, Warkne PC, Molnar P, Lapin GD, Mikhael MA. Effect of hyperosmotic blood-brain barrier disruption on transcapillary transport in canine brain tumors. J Neurosurg. 1990; 72: 441–9. doi: 10.3171/jns.1990.72.3.0441.
[35]  Neuwelt EA, Goldman DL, Dahlborg SA, Crossen J, Ramsey F, Roman Goldstein S, et al. Primary CNS lymphoma treated with osmotic blood-brain barrier disruption: prolonged survival and preservation of cognitive function. J Clin Oncol. 1991; 9: 1580–90. doi: 10.1200/JCO.1991.9.9.1580.
[36]  Zünkeler B, RE C, Olson J, Blasberg RG, DeVroom H, Lutz RJ, et al. Quantification and pharmacokinetics of blood-brain barrier disruption in humans. J Neurosurg. 1996; 85: 1056–65. doi: 10.3171/jns.1996.85.6.1056.
[37]  Burkhardt JK, Riina H, Shin BJ, Christos P, Kesavabhotla K, Hofstetter CP, et al. Intra-arterial delivery of bevacizumab after blood-brain barrier disruption for the treatment of recurrent glioblastoma: progression-free survival and overall survival. World Neurosurg. 2012; 77: 130–4. doi: 10.1016/j.wneu.2011.05.056.
[38]  Bartus RT. The blood-brain barrier as a target for pharmacological modulation. Curr Opin Drug Discov Dev. 1999; 2: 152–67.
[39]  Gumerlock MK, Neuwelt EA. The effect of anesthesia on osmotic blood-brain barrier disruption. Neurosurgery. 1990; 26: 268–77. doi: 10.1227/00006123-199002000-00014.
[40]  Kroll RA, Neuwelt EA. Outwitting the blood-brain barrier for therapeutic purposes: osmotic opening and other means. Neurosurgery. 1998; 42: 1083–100. doi: 10.1097/00006123-199805000-00082.
[41]  Raymond JJ, Robertson DM, Dinsdale HB. Pharmacological modification of bradykinin-induced breakdown of the blood-brain barrier. Neurol Sci. 1986; 13: 214–22. doi: 10.1017/S0317167100036301.
[42]  Inamura T, Black KL. Bradykinin selectively opens blood-tumor barrier in experimental brain tumors, J. Cerebr. Blood Flow Metabol. 1994; 14: 862–70.