22 Intra-arterial Chemotherapy


 



Abstract


Intra-arterial chemotherapy (IAC) for malignant gliomas is an attractive treatment strategy that utilizes the selective nature of arterial catheterization to minimize nontarget drug toxicity allowing for higher local drug concentration. However, early case reports and randomized controlled trials (RCTs) failed to show discernable survival benefit. The interval development of more navigable catheters, fine-tuning of tumor-specific chemotherapy agents, and innovative blood-brain barrier disruption techniques have enabled increased selectivity for treatment. Early reports are showing modest but persistent survival benefits of adjunct IAC for malignancies of the intracranial compartments, as well as the head and neck. Reviewed here is the evidence of IAC in the treatment of recurrent malignant gliomas, primary central nervous system (CNS) lymphoma, lacrimal gland malignancies, and squamous cell carcinoma of the head and neck. RCTs have evaluated a select few chemotherapy agents for malignant gliomas and squamous cell carcinomas of the head and neck and the remainder of the evidence rests on case series and case reports. However, many phase III trials for IAC are open and recruiting.




22 Intra-arterial Chemotherapy



22.1 Goals




  1. Review the current clinical applications of intra-arterial chemotherapy (IAC) beyond retinoblastoma.



  2. Critically analyze the literature on IAC on gliomas, squamous cell carcinomas of the head and neck, primary central nervous system (CNS) lymphoma, intracranial metastases, and adenoid cystic carcinoma (ACC) of the lacrimal gland.



  3. Briefly review the variety of applications of IAC in the treatment of uncommon intracranial tumors and experimental efforts to expand the use of IAC.



22.2 Case Example



22.2.1 History of Present Illness


A 43-year-old woman who initially presented approximately 1 year after noticing a small nodule over the right superolateral globe which eventually was associated with mild right temporal discomfort. The condition was initially treated as a prolapsed lacrimal gland that was repositioned surgically. Biopsy at the time demonstrated mild chronic inflammation. Follow-up imaging performed a year later showed progressive enlargement. Excisional biopsy of the nodule demonstrated ACC. Follow-up imaging noted persistent asymmetry of the right lacrimal gland but no abnormally enhancing lesion.


Past medical history: Left arm and ankle fracture after car accident.


Past surgical history: Laparoscopic cholecystectomy 2011, nasal septum repair 1988.


Family history: Father died of stomach cancer; mother alive at age 66 with chronic obstructive pulmonary disease (COPD).


Brother and sister in their 40 s are in good health.


Social history: University professor. Denies smoking or other chemical exposures. Social drinker.


Review of systems: Some blurry vision and eye pain.


Neurological examination: CN II-IX, XI, XII intact, PERRL, EOMI, 5/5 motor strength and sensation.


Imaging studies: See Fig. 22.1 and Fig. 22.2.



22.2.2 Treatment Plan


There is a significant reduction in cause-specific mortality at 5 years with intra-arterial cytoreductive chemotherapy (IACC). Patient was admitted for neoadjuvant IAC with cisplatin and Adriamycin on day 1 followed by IACC with Adriamycin for another 2 days. She was premedicated with decadron, Zofran, and Ativan, and started on Neulasta on treatment day 2.

Fig. 22.1 Coronal noncontrast computed tomography (CT) of the orbits (a) demonstrates asymmetrically enlarged soft tissue density of the right lacrimal gland. Corresponding coronal T2 image of the orbits (b) demonstrates heterogenous T2 hypertense asymmetric soft tissue abnormality of the right lacrimal gland. (Photo credit: Priyank Khandelwal, MD.)
Fig. 22.2 Frontal (a) and lateral (b) views of a digital subtraction angiogram of the right external carotid artery prior to treatment with intra-arterial chemotherapy of a right lacrimal gland adenocys-tic carcinoma. Lacrimal branches of the right internal maxillary artery were superselected for intra-arterial chemotherapy administration. There is no significant tumor blush of the right lacrimal region. Control frontal (c) and lateral (d) angiography of the right ECA demonstrates a similar appearance of the parent vessel. (Photo credit: Priyank Khandelwal, MD.)


22.2.3 Follow-up


Patient initially complained of right-sided tinnitus and tingling of the right head and extremities which dissipated by day 3. She had no hematologic alterations, cardiovascular changes, or nephrotoxicity. On her last clinic visit 5 years posttreatment, she is doing well. She complains of occasional dry eyes but denies blurry vision, headache, or local symptomatology.



22.3 Case Summary


What are the circumstances when IAC might be a reasonable treatment option?



22.3.1 Adenoid Cystic Carcinoma of the Lacrimal Gland


Adenoid cystic carcinoma (ACC) is a rare tumor of the oral and maxillofacial regions predominately found in the minor salivary glands, 1 but can occur in any secretory gland including the breast, cervix, esophagus, lungs, prostate, and lacrimal gland. 2 Lacrimal gland ACC carries a poor prognosis despite disfiguring wide local excision because of its propensity for perineural spread and distant temporal recurrence. 3 The current standard of care is radical orbitectomy, but eye-sparing procedures have been gaining support in the literature because of similar survival rates to radical surgical excision. 4


Recurrence most commonly occurs locally. 5 For this reason, adjuvant radiation therapy and chemotherapy on top of surgical excision is considered the most promising treatment option. 6 A case report 7 and larger case series 8 have reported improved overall survival and decreased recurrence with IAC for lacrimal gland ACC followed by orbital exenteration, chemoradiation, and IV chemotherapy. Patients who completed treatment had a significantly higher 10-year disease-free survival when compared to the group undergoing conventional treatment (100 vs. 14.3%); however, this group excluded those who were not given IAC due to sacrifice of the lacrimal artery from excisional biopsy, precluding straightforward catheterization. IAC resulted in downstaging primary tumors with intracranial extension or temporal fossa involvement to disease limited to the intraorbital compartment. One major adverse event occurred in a patient who was treated through the ICA and sustained central retinal artery occlusion and complete vision loss prior to exenteration.


A smaller case series of four patients who underwent IAC followed by resection reported significant adverse effects including eye lid and globe necrosis, facial palsy, difficulty chewing, facial swelling, and neutropenia. 9 Three patients had total exenteration, and at follow-up (range 48-69 mo), there was no evidence of recurrence of metastases. The fourth patient had lacrimal gland resection alone and at 54 months was found to have local recurrence for which total exenteration was recommended.























































































































































Table 22.1 Untreated malignant gliomas: single IA agent series and trials
Study Year IA agent N CR PR MS (mo)
Yamashita 1983 ACNU 17 (GBM) NR NR 12.5 IA, 9 IV
Greenberg 1984 BCNU 36 (12 Grade III–IV, 24 grade I–II) NR NR Prior surgery 13.5 Prior RT 6.5
Hochberg 1985 BCNU 79 (39 New) NR NR 12.5–13.5
Bashir 1988 BCNU 28 (Grade III–IV) 22.0 22.0 9.2
Greenberg 1988 BUdR 18 (GBM and AA) 17.0 28.0 22
Wolpert 1988 BCNU 10 NR NR No outcomes discussed
Clayman 1989 BCNU 15 (12 New) 33.3 6.7 19.2
Roosen 1989 ACNU 50 (42 New) NR 57.1 14.2
Fauchon 1990 HeCNU 40 (33 GBM, 6 AA) 5.0 0.1 10.5 (GBM only)
Dropcho 1992 Cisplatin 22 (13 GBM, 9 AA) NR 0.2 15.7
Mortimer 1992 Cisplatin 22 5.0 23.0 9.6–10.8
Vega 1992 ACNU 22 (4 GBM, 18 AA) NR NR 6
Shapiro 1992 BCNU IA vs. IV 315 (94 GBM IV, 111 GBM IA) NR NR 11.2 (IA), 14 (IV)
Chauveinc 1996 ACNU 27 (10 GBM, 17 AA) NR NR 10
Kochii 2000 ACNU IA vs. IV 82 NR NR 14.8 (IA), 14 (IV)
Imbesi 2006 ACNU IA vs. IV 43 NR NR 17 (IA), 20 (IV)
Abbreviations: AA, anaplastic astrocytoma; ACNU, nimustine; BCNU, carmustine; BUdR, bromodeoxyuridine; CR, complete response; GBM, glioblastoma multiforme; HeCNU, 1-(2-hydroxyethyl)-3-(2 chloroethyl)-3-nitrosourea; IA, intra-arterial; IV, intravenous; MS, median survival; NR, not recorded; PR, partial response—generally > 50% decrease in tumor volume; RT, radiotherapy.

Stage I and II disease or tumors less than 4 cm without ex-traparenchymal extension do well with globe preservation surgery and radiation, with local recurrence observed in those with greater residual tumor burden. 10 Given the high morbidity of orbital exenteration, globe-sparing surgery is gaining popularity with adjuvant proton radiation therapy or concurrent chemoradiation. 11 IAC may play a growing role in reducing microscopic tumor burden.



22.3.2 Intracranial Gliomas


For newly diagnosed malignant gliomas, the standard of treatment entails surgical resection with adjuvant chemoradiation, 12 , 13 , 14 yielding an overall survival of 15 months. 15 , 16 Methylated 0[6]-methylguanine-DNA methyltransferase (MGMT)-positive histology confers a better prognosis with overall survival of 23.4 months. 17 IAC is currently utilized either in clinical trials or as off-label salvage therapy. 18


There are four phase III randomized controlled trials (RCTs) comparing IA with systemic chemotherapy 19 , 20 , 21 , 22 in patients with newly diagnosed glioblastoma multiforme. All patients underwent surgical resection followed by radiation therapy with IA carmustine (BCNU), 19 , 21 nimustine (ACNU), 20 and carbo-platin and ACNU. 22 These initial chemotherapeutic agents were chosen for their lipophilic properties. 23 IA therapy was administered through the internal carotid and vertebral arteries, and in some cases the dose was divided between arteries, if the tumor crossed arterial territories. 21 Three of the four studies were terminated at interim analysis. The trials’ cumulative 469 patients successfully randomized showed no survival benefit for IAC.


Thousands of patients have been enrolled in phase I and II clinical trials testing the safety and efficacy of IAC for newly diagnosed as well as recurrent grade III to IV gliomas. Early small case series for newly diagnosed malignant gliomas focused on nitrosoureas (ACNU, BCNU, and hydroxyethyl-chloroethyl nitrosourea [HeCNU]) on the basis that lipophilic agents would facilitate passage across the blood-brain barrier. Leukoencephal-opathy and ocular toxicity were common side effects, prompting combination therapy. Median survival ranges from 5 to 22 months (Table 22.1, ▶ Table 22.2).


With recurrent tumors, overall survival is 25 weeks. 24 One multi-institutional RCT found a statistically significant survival benefit of IV PCNU over IA cisplatin (13 vs. 10 mo). 25 Although IV PCNU was associated with greater hematotoxicity, IA cisplatin led to greater renal toxicity, ototoxicity, and encephalopathy.


Similar to the literature on newly diagnosed malignant gliomas, early trials in the treatment of recurrent tumors ( Table 22.3) initially focused on nitrosoureas before moving toward platinum-based agents (cisplatin, carboplatin). Reports of ocular, hematologic, and neurologic toxicities likely spurred interest in immunologic agents and combination IA, IV, and PO therapy. In the recurrent tumor IA literature, there are more reports of these new agents as well as the use of hydrophilic compounds with adjuvant blood-brain barrier disruption (BBBD). 26 Despite greater patient volume and outcomes data, conclusions are difficult to make on the whole because of treatment and patient heterogeneity. Many studies include not only patients with various prior surgical, medical, and radiation histories but also add adjuvant or concurrent radiation in the treatment arm along with multiagent therapy (▶ Table 22.4).


Poor support to IA therapy may be related to the heterogeneity of the underlying pathology, toxicity of earlier chemotherapeutic agents, and poor patient selection. MGMT gene expression confers a particular benefit from temozolomide and may be used for patient selection for super selective intra-arterial chemotherapy (SSIAC) trials. 27 , 28 Other prognostic factors for survival include young age, good clinical performance status, extensive surgical resection, seizures, and histologic features. 29 In the highly selected populations undergoing IAC, it is difficult to surmise which patient characteristics predict a good response. In one series of 41 patients receiving IAC, 80% of which were glioblastomas (GBMs), half of the tumors were avascular and the other half very hypervascular on angiography. Hypervascularity was a statistically significant predictor of decreased survival. 30 Imaging related predictors of increased survival included small enhancing tumor volume, avascularity, and longer time to recurrence which was defined as clinical deterioration or more than 25% increase in tumor area on magnetic resonance imaging (MRI) (fluid-attenuated inversion recovery [FLAIR] hyperintensity or enhancement). Small tumor volume and avascularity are thought to be related as more aggressive tumors tend to more vascular because of increased micro-vessel density 31 and tumoral angiogenesis. 32























































































Table 22.2 Untreated malignant gliomas: multi-IA agent series and trials
Study Year IA agent N CR PR MS (mo)
West 1983 BCNU IV vincristine, procarbazine 16 0.0 80.0 17.1
Madajewicz 1991 Cisplatin, etoposide 48 (13 new) 5.0 37.0 14 (GBM)
Watne 1991 BCNU IV vincristine, PO procarbazine 79 (19 AA, 60 GBM) NR NR 30 overall 10 GBM
Bobo 1992 BCNU and cisplatin 39 (21 AA, 18 GBM) 16.7 27.8 12.5
Watne 1993 BCNU IV vincristine, PO proarbazine 173 (35 AA, 38 GBM) NR NR GBM 10, AA 57
Nakagawa 1994 Cisplatin, etoposide 7 (4 GBM, 3 AA) 25 (GBM) 33 (AA) 25 (GBM), 33 (AA) 5
Madajewicz 2000 Cisplatin, etoposide 83 (63 GBM, 20 AA) 5.6 (all), 8.9 (GBM) 42 (all), 41 (GBM) 20 before RT 7 concurrent RT
Silvani 2002 IA carboplatin and ACNU or IV cisplatin and BCNU 30 0.0 21 (IA), 33 (IV) 18.3 (IA), 18.6 (IV)
Abbreviations: AA, anaplastic astrocytoma; ACNU, nimustine; BCNU, carmustine; CR, complete response; GBM, glioblastoma multiforme; IA, intraarterial; IV, intravenous; MS, median survival; NR, not recorded; PR, partial response—generally > 50% decrease in tumor volume; RT, radiotherapy.

Major hurdles in selective drug delivery include the an-gioarchitecture of GBMs which derive vascular supply from adjacent vascular territories or even the contralateral hemisphere. 23 Postcontrast enhancement on MRI typically underestimates the vascular territory involved, a finding overcompen-sated by T2 signal abnormalities. 33 Using FLAIR hyperintensity, 66% of tumors were supplied by both anterior and posterior territories and 10% were supplied by three arterial territories. This suggests that catheterization of a single intracranial artery is insufficient, and effective treatment likely involves superse-lection of branches in different vascular territories.


Most recent SSIAC case series have been performed in those with recurrent high-grade gliomas. 34 , 35 , 36 One group has published phase I and II clinical trials employing SSIAC with bevaci-zumab for recurrent glioblastoma. 34 , 35 IAC was administered with concomitant balloon occlusion of downstream parent vessels. Follow-up MRI at 1 month also demonstrated decrease in tumor volume as measured by FLAIR hyperintensity, perfusion volume, and enhancement. IAC conferred a median progression-free (PFS) and overall (MOS) survival of 10 and 8.8 months, respectively, which was comparable to prior studies using IV bevacizumab where medial PFS and MOS ranged 3.7-4.2 and 7.2-9.2 months, respectively. 37 , 38


Qureshi et al reported a case series of 12 GBM patients treated with SSIAC carboplatin and nonapeptide H-Arg-Pro-Hyp-Gly-Thi-Ser-Pro-Tyr(Me)-ip(CH2NH)-Arg-OH (RMP-7) for BBBD (discussed later). All were cases of recurrent GBM after gross total resection and chemoradiation. In the six patients with GBM for whom follow-up data was available, one-half demonstrated decreased tumor volume on MRI, defined as enhancement on Tl-weighted images, at a median follow-up of 4.9 months. One-year survival for GBM patients was 45% (5 out of ll). 36


SSIAC decreases nontarget drug delivery while increasing the tumor tissue dose, particularly those supplied by small distal branches. Superselective catheterization of the middle cerebral artery (MCA) was associated with fewer ocular toxicities. 39 In later studies employing SSIAC, there is a notable decrease in the incidence of seizures, leukoencephalopathy, and hematologic toxicities. 34 , 35 , 36 Interestingly, those receiving intraprocedural RMP-7 for BBBD had fewer neurological complications, though not statistically significant. 36


The advantage of SSIAC is direct administration to the tumor bed to achieve high local concentrations. To decrease streaming, pulsatile dosing and spatial dose fractionation was used to deliver a more tailored dose for the territory supplied by a given artery. This accounts for individual variations in the circle of Willis so that dosing is based on arterial territory volume and not body weight or body surface area as a way to decrease neurologic complications from administering chemotherapy to normal tissues. 18


Advances in the understanding of drug delivery have informed clinical trials. Initially postcontrast enhancement on MRI of the brain was used to indicate blood-brain barrier breakdown and support the use of IAC. 40 While degree of enhancement does reflect tissue uptake of chemotherapeutic drugs, 26 animal studies comparing MRI enhancement with drug uptake in tumor versus normal tissues have demonstrated unacceptably high doses in normal brain tissue as well as systemically. 41 This has fueled research in local disruption of the blood-brain barrier to facilitate drug delivery.























































































































































































Table 22.3 Recurrent malignant gliomas: single IA agent series and trials
Study Year IA agent N CR PR MS (mo)
Lehane 1982 Cisplatin 10 (Grade III–IV) 0.0 80.0 4
Feun 1984 Cisplatin 35 (18 GBM, 3 AA) 0.0 28.6 (Grade II–IV) NR
Greenberg 1984 BCNU 36 (31 Grade III–IV) 8.0 62–67 13–13.5
Hochberg 1985 BCNU 79 (25 Recurrent) NR NR 13.5
Feun 1987 Etoposide 28 (13 GBM, 1 AA) 0.0 0.0 NR
Johnson 1987 BCNU 20 (18 GBM) 0.0 10.0 8.7
Mahaley 1989 Cisplatin 40 (20 GBM, 14 AA) 0.0 18 (Overall) 4.1
Newton 1989 Cisplatin 12 (9 GBM or AA) 0.0 8.0 NR
Calvo 1989 Cisplatin 12 (8 GBM) 41.7 50.0 10
Roosen 1989 ACNU 50 (7 Recurrent) NR NR 6.1
Poisson 1990 HECNU 53 (30 GBM, 10 AA) 17 (GBM) 40 (AA) 13 (GBM) 10 (AA) 4.5 (GBM) 18 (AA)
Saris 1991 Cisplatin 10 (7 GBM, 3 AA) 10.0 0.0 NR
Bradac 1992 BCNU 17 (12 GBM, 5 AA) 0.0 29 (All AA) 6.7 (AA) 6 (GBM)
Stewart 1992 Carboplatin 15 (6 GBM, 1 grade II–III, 2 grade II) 0.0 0 (GBM) 33 (Grade II–III) 1.5
Vega 1992 ACNU 18 (6 GBM, 12 AA) 0 GBM 25 (AA) 33 (GBM) 17 (AA) 8
Hiesiger 1995 IA cisplatin vs. IV PCNU 311 (174 GBM) NR NR 10 (IA) 13 (IV)
Chow 2000 Carboplatin with cereport 46 (32 GBM, 9 AA) NR NR NR (7.3–11.6)
Qureshi 2001 Carboplatin with RMP-7 24 (12 GBM, 6 AA) 8 (GBM) 8 (GBM) 17 (AA) 7 (GBM) 25.5 (AA)
Boockvar 2011 Bevacizumab and mannitol vs. IV bevacizumab 30 (26 GBM, 2 AA) NR NR NR
Burkhardt 2012 IA bevacizumab and mannitol IV bevacizumab 14 GBM 0.0 57.0 10
Abbreviations: AA, anaplastic astrocytoma; ACNU, nimustine; BCNU, carmustine; CR, complete response; GBM, glioblastoma multiforme; IA, intraarterial; IV, intravenous; MS, median survival; NR, not recorded; PR, partial response—generally > 50% decrease in tumor volume; RT, radiotherapy.

Clinical trials employing IAC with BBBD are accruing patients and results are promising. The most commonly employed agent is hyperosmolar mannitol infused right before chemotherapy administration to induce osmotic shrinkage of the endothelial cells lining the capillaries to promote separation of tight junctions. 42 An alternative agent is lobradimil (also known as RMP-7 or Cereport), a bradykinin derivative that increases vascular permeability, and regadenoson an adenosine agonist causing transient BBBD. 43


In addition to superselective catheterization, other strategies for efficient drug delivery include flow arrest to maximize drug extraction and minimize drug streaming. High cerebral blood flow and low drug concentrations hinder tissue uptake. Streaming can be overcome with injections of drug volumes greater than 20% of the background blood flow rate, injecting during diastole, or using catheters with side ports. 44 , 45 , 46 These delivery pharmacokinetics are studied using the Dedrick model of blood flow. 47


Convection-enhanced drug delivery is another catheter-based strategy where catheters are directly implanted into the tumor bed. Drugs are pressure infused at a set concentration, rate, and duration across the leaky cytoarchitecture of growing tumor cells 48 and across pre-existing tracts of white matter edema. 49 Small phase I and II trials have experimented with monoclonal antibodies, liposomal vectors packaged for gene therapy, antisense oligonucleotides, and exotoxins. 50 However, early phase III trials were not able to demonstrate a survival benefit, 51 although PFS was significantly increased in the treatment arm, 17.7 versus 11.4 weeks.


Riina et al have reported the use of SSIAC with bevacizumab and mannitol for a malignant glioma in 2009 and for a malignant brainstem glioma in 2010 with decreased enhancement seen on follow-up MRI in both cases. 52 , 53 Numerous clinical trials have also described IAC in the treatment of diffuse intrinsic pontine gliomas (DIPGs), 54 esthesioneuroblastoma, 55 anaplastic oligodendrogliomas, 56 germ cell tumors, 57 cerebral metastatic disease. However, these treatments are entirely experimental.







































































Table 22.4 Recurrent malignant gliomas: multi-IA agent series and trials
Study Year IA agent N CR PR MS (mo)
Neuwelt 1991 IA MTX, IV mannitol post-RT vs. pre-RT 30 70.0 30.0 17.8 post-RT vs. 44.5 pre-RT
Doolittle 2000 IA carboplatin IV mannitol, cyclophosphamide, etoposide 221 (53 PCNSL) 75.5 15.1 NR
Fortin 2005 IA carboplatin IV etoposide and cyclophosphamide 31 (8 PCNSL) 37.5 62.5 Not reached
Sonoda 2007 IA ACNU and RT 63 75.0 25.0 39
MacNealy 2008 IA MTX IV etoposide, cyclophosphamide, mannitol Primary meningeal lymphoma case report
Angelov 2009 IA MTX, IV mannitol 149 57.8 24.2 37
Abbreviations: AA, anaplastic astrocytoma; ACNU, nimustine; BCNU, carmustine; CR, complete response; GBM, glioblastoma multiforme; IA, intraarterial; IV, intravenous; MS, median survival; MTX, methotrexate; NR, not recorded; PCNSL, primary CNS lymphoma; PR, partial response— generally > 50% decrease in tumor volume; RT, radiotherapy.

This limited availability has led to international controversy in the media for rare malignancies for which there is no accepted treatment. 58

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

May 4, 2022 | Posted by in NEUROSURGERY | Comments Off on 22 Intra-arterial Chemotherapy

Full access? Get Clinical Tree

Get Clinical Tree app for offline access