Summary and Commentary on Previously Published Neuropathology Guideline
As noted above this review is an update to the previously published guidelines for progressive glioblastoma by Brat et al.[8] It is useful to briefly review the questions and results from that paper.
The first topic addressed the diagnostic considerations in reporting progressive glioblastoma. A level III recommendation that the pathologist consider the patient’s previous diagnosis and treatment, as well as the current clinical and neuroimaging features that led to a second biopsy or resection. In the setting of prior radiation and chemotherapy, it was recommended the pathologist adhere to strict histologic criteria for microvascular proliferation and necrosis in order to establish a diagnosis of a glioblastoma.
For patients undergoing biopsy or neurosurgical resection at the time of radiologic or clinical progression, reporting the presence and extent of progressive neoplasm as well as the presence and extent of necrosis within the pathologic material examined was recommended.[8] This recommendation continues to be supported in this update and with recent publications adding further credence to these as described below.
There is often a combination of radiation necrosis and progressive glioma in biopsy and resection specimens and it can be difficult to histologically assess disease status. Additional manuscripts published since the last version of this guideline refine the understanding of this issue. This was particularly emphasized by the study conducted by Holdhoff, et al., in which only “marginal agreement” by Fleiss’ kappa statistics was observed when 48 pathologists (92% of whom were neuropathologists) reviewed 13 cases of suspected recurrence of glioblastoma. [14] Much of this may be due to differential understanding of the terminology used in this this study. The terminology of “active tumor”, “inactive tumor”, and “treatment effect” are not well established in the literature[14] yet this is commonly used terminology.
Multiple studies have aimed to determine what features give the greatest prognostic information on second resection specimen, methodology and results have varied. Azoulay et al., Dalle Ore et al., Hu et al., and Woodworth et al., show that the majority of re-resection specimens demonstrate a mixture of tumor and therapy related effects (ranging from 49-85% of specimens), with only a minority demonstrating absence of active residual tumor (ranging from 5-29% of patients).[9; 10; 15; 16] Azoulay et al. and Woodworth et al. demonstrated that patients in whom the re-resection specimen demonstrated only therapy related changes without active tumor had an increased survival.[15; 16] Hu et al. demonstrated a significant association between percent tumor and overall survival,[10] however Bagley et al. and Dalle Ore et al. were not able to reproduce these findings[5; 9] (See Table I). Another suggested marker of prognosis present in re-resection specimens has been the proliferative index as assessed by Ki-67 / MIB-1 immunohistochemistry. Okita et al. were able to demonstrate that MIB-1 indices significantly correlated with overall survival in a multivariate analysis (p=0.004)[17] (See Table II). Thus, it is recommended that the percentage of viable tumor and the percentage of radiation necrosis is documented. If available the proliferative index as assessed by MIB-1 immunohistochemistry may also be informative, but it is not felt there is enough evidence at this time to include this as part of the recommendation (See Table II).
The second question in the prior version of this guideline addressed what ancillary studies are most useful in differentiating progression from treatment effect. Immunohistochemistry, including Ki-67, IDH, p53, and WT1, and genetic studies, specifically EGFR amplification or gain of chromosome 7, were selectively recommended for distinguishing neoplastic cells from atypical reactive cells in progressive glioblastoma.[8] This update does not alter this recommendation but numerous additional studies that have been published since then and have better elucidated the impact some of these features have on the behavior of infiltrating gliomas. Indeed, this was reflected in the World Health Organization 2016 Classification of Tumors of the Central Nervous System[12] which incorporates selected immunohistochemical and genetic features in the classification system. Thus, while the prior review focused on using these markers to determine the presence and in some cases quantity of tumor present, several of these studies are now indicated for classification and prognostic uses as well.
The integration of ancillary studies is expected to expand with the publication of the next World Health Organization Classification of Tumors of the Central Nervous System with some of the expected updates already published in the form of cIMPACT-NOW update recommendations.[18] For the current review of progressive glioblastoma, criteria of the World Health Organization classification with the addition of the published cIMPACT-NOW update 3 recommendations will be used, since they represent a recent and updated international standard for classifying and grading.[12; 18] An additional cIMPACT-NOW update 5 was published after the designated time interval of the search for this guideline and the information in this document are not used to formulate the updated recommendations.[19]
It is of value to briefly expand on the evolution of the immunohistochemical and molecular markers alluded to in the second question of the prior guideline as it provides background on the new questions asked in this update as noted below. The 2016 WHO classification divides the diffuse gliomas into IDH-wildtype astrocytomas, IDH-mutant astrocytomas, IDH-mutant and 1p/19q codelted oligodendrogliomas, and H3K27M-mutant diffuse midline glioma. A 3-tiered grading system is used in grading diffuse astrocytic gliomas, and a 2-tiered grading system of oligodendrogliomas.[12]
IDH-wildtype diffuse astrocytomas tend to arise in an older patient population with large majority of them presenting as de novo glioblastomas.[20] As such IDH-wildtype diffuse astrocytoma, WHO grade II and anaplastic astrocytoma (WHO grade III) are recognized as provisional entities in the 2016 WHO and multiple studies have concluded that a substantial subset of these demonstrate an aggressive clinical course most akin to IDH-wildtype glioblastoma, WHO grade IV. Recently the cIMPACT-NOW update 3 addressed this by recommending that IDH-wildtype infiltrating astrocytomas that would histologically be classified as WHO grade II or III which carry and EGFR amplification, combined whole chromosome 7 gain and whole chromosome 10 loss, or a TERT promoter mutation be given an integrated diagnosis of “diffuse astrocytic glioma, IDH-wildtype, with molecular features of glioblastoma, WHO grade IV.” It is critical to note that other IDH-wildtype glial tumors that may enter the differential have been reported to harbor TERT promoter mutations and thus histologic examination of the specimen remains critical.[18]
IDH mutant diffuse astrocytomas tend to arise in younger patient population and tend to present as either a diffuse astrocytoma WHO grade II or anaplastic astrocytoma WHO grade III. They are associated with TP53 mutations and ATRX alterations and are relatively slowly progressive.[12; 20; 21]
Diffuse midline glioma (H3K27-mutant) and H3 G34 mutant diffuse gliomas do not have IDH-mutations and occur predominantly in childhood and adolescents and are associated with an aggressive clinical course. Diffuse midline gliomas (H3K27M-mutant) are by definition WHO grade IV. While the 2016 World Health Organization Classification of Tumors of the Central Nervous System did not provide a separate classification or grade for H3 G34-mutant diffuse glioma, this mutation in a diffuse glioma indicates a high-grade biology with only modestly longer survivals than other IDH-wildtype glioblastomas. In most cases the assessment of prognostic genetic markers will have been performed on the initial specimen, however, if they were not performed previously they should be performed at the time of progression. Specific alterations that have current diagnostic, prognostic, or therapeutic implications in diffuse astrocytic gliomas include IDH1 and IDH2 mutations, EGFR amplification, whole chromosome 7 gain with concurrent whole chromosome 10 loss, TERT promoter mutation, CDKN2A homozygous deletion, 1p/19q co-deletion, and MGMT promoter methylation status.[8; 12; 18]
Question: For adult patients with progressive glioblastoma does testing for Isocitrate Dehydrogenase (IDH) 1 or 2 mutations provide new additional management or prognostic information beyond that derived from the tumor at initial presentation?
Study selection and Characteristics:
Four studies focused on IDH mutation status in primary and progressive glioblastoma were uncovered in the initial screen.[22; 23] Only two of these papers focused on the use of IDH mutations in recurrent glioblastoma. Two additional papers were identified from relevant review articles that addressed maintenance of IDH mutations during glioma progression[24; 25] (Table IV).
Results of individual studies, discussion of study limitations and risk of bias
Mutations in IDH are frequent in lower grade astrocytomas as well as glioblastomas that progress from these lower grade precursor lesions. IDH mutations most commonly occur in IDH1 as a substitution of histidine at R132, which accounts for approximately 90% of all IDH mutations.[12; 23] The IDH1 R132H mutation can be detected by either immunohistochemical or molecular methods. Approximately 10% of IDH mutations are due to alternate mutations in IDH1 or on IDH2, including IDH1 R132C, IDH1 R132G, IDH1 R132S and IDH1 R132L and IDH2 R172K,[26] which can currently only be detected by molecular testing. IDH mutations tend to occur early in glioma development and they are retained upon recurrence, even after therapy.[24; 25; 27] IDH mutations contribute to glioma development through overproduction of the oncometabolite 2-hydroxyglutarate leading to induction of the HIF-1 pathway and genome wide histone and DNA methylation alterations.[12; 28-30] Testing for IDH1/2 mutations may be useful in the context of progressive glioblastoma when attempting to determine if residual disease is present in a resection specimen and to assess the approximate percentage of tumor.[8; 22] It is also feasible to use IDH mutation testing to aid in frozen section diagnosis, however, the turnaround time is approximately 1 hour and may not be practical for all laboratories.[22] It should be noted that infiltrating gliomas, and glioblastomas are heterogeneous tumors and rare instances of alterations in IDH status from that seen in the original lesion have been noted, usually a loss of the IDH mutation[31–33](See Table III).
Synthesis
Due to the early occurrence and conserved nature of IDH mutations repeat testing is not necessary if the tumor is histologically similar to the primary tumor and the patient’s clinical course is as expected.
Question: For adult patients with progressive glioblastoma does repeat testing for MGMT promoter methylation provide new or additional management or prognostic information beyond that derived from the tumor at initial presentation and what methods of detection are optimal?
Study selection and Characteristics
Thirteen papers were uncovered in the screening process that discussed alterations in MGMT promoter methylation status in progressive/recurrent glioblastoma. Eleven were selected for inclusion as 1 was a review article and 1 paper only had four paired tumors and did not specifically note if MGMT promoter methylation status was retained in those four cases.
Results of individual studies, discussion of study limitations and risk of bias
Temozolomide, a chemotherapeutic agent that is part of the standard therapy for glioblastoma, and other alkylating agents cause DNA crosslinking through alkylation of the O6 position guanine. These alkyl adducts are removed by O6-methylguanine-DNA methyltransferase (MGMT). Methylation of the MGMT promoter is one of the major mechanisms for MGMT regulation and leads to transcriptional silencing. Thus, glioblastomas with MGMT promoter methylation and thus lower expression of MGMT would be expected to have better response to alkylating agents.[13; 34] MGMT promoter methylation has become a clinically relevant prognostic and predictive marker in patients with glioblastoma treated with temozolomide or other alkylating agents and is associated with a statistically significant improvement in progression free survival and overall survival in patients receiving standard therapy.[34; 35]
MGMT promoter methylation status is fairly consistent from original tumor presentation to recurrence, with no status change in approximately 66–82% of glioblastomas.[17; 36] No significant intra-tumor heterogeneity of MGMT promoter methylation status has been found.[36] Brandes et al. demonstrated that overall survival correlated with MGMT promoter methylation status determined at the primary surgery but not at recurrence and Okita et al. found no correlation between MGMT promoter methylation status at recurrence and survival time or progression free survival.[17; 36] A few earlier studies did show correlation with MGMT promoter methylation at recurrence and chemoresistance and survival,[37] however, the majority of the data suggests that repeat testing is not needed.
Multiple methods of MGMT promoter methylation are available, including pyrosequencing, quantitative real-time methylation specific PCR, and methylation specific PCR. Recent studies have compared the multiple methods and tried to determine optimal stratification. The best performance was seen using a 3 tiered system of unmethylated, low level methylation, and high level methylation using either pyrosequencing or methylation specific PCR, with pyrosequencing being the preferred method.[34; 38-41]
An immunohistochemical stain for MGMT protein expression is also available, however MGMT protein expression can be upregulated by glucocorticoids, chemotherapy, and radiotherapy and thus my not reflect true MGMT status. It is also prone to high inter-observer variability and shows inconsistent correlation with clinical outcomes[38; 39; 42-44](See Table IV).
Synthesis
Repeat MGMT promoter methylation testing does not need to be repeated upon recurrence. Either pyrosequencing or methylation specific PCR can be used to assess MGMT promoter methylation, with pyrosequencing being the preferred method. Immunohistochemical testing for MGMT protein expression is not recommended for clinical use.
Question: For adult patients with progressive glioblastoma does EGFR amplification or mutation testing provide management or prognostic information beyond that provided by histologic analysis and if performed on previous tissue samples, does it need to be repeated?
Study selection and Characteristics
Twenty articles were uncovered in the screening process of which 9 articles were selected for inclusion in this review and 1 additional article was identified from the references of the included articles. Reasons for exclusion from the included publications included animal or in-vitro studies, phase 1 clinical trials that were ended early or in which therapeutic outcomes were not discussed, review articles or single case studies, or did not address pathology (studies with only imaging data).
Results of individual studies, discussion of study limitations and risk of bias
EGFR is the most commonly amplified and overexpressed proto-oncogene in glioblastoma, with amplification present in approximately 40-50% of glioblastomas.[45–47] Amplification is primarily seen in de novo glioblastomas[45] and appears to be mutually exclusive of IDH mutations.[30; 48] EGFR is located on the short arm of chromosome 7 and encodes a cell-surface receptor tyrosine kinase. EGFR activation initiates signal transduction through several major pathways including RAS-MAPK and PI3K-AKT signal transduction cascades leading to increased DNA transcription, anti‐apoptosis, angiogenesis and cellular proliferation.[49–51] EGFR overexpression was found to contribute to gliomagenesis and poor survival in patients with glioblastoma.[52]
Mutations of EGFR are also seen in IDH-wildtype glioblastoma and appear to occur exclusively in the setting of EGFR amplified glioblastomas. EGFRvIII is the most common mutation in glioblastoma and results in the creation of a tumor-specific antigen that is detectable in 23–33% of IDH-wildtype glioblastomas,[53; 54] and in approximately 50% of EGFR amplified glioblastomas.[46; 55; 56] EGFRvIII is the result of a deletion of EGFR exons 2–7, which generates a constitutively active tyrosine kinase with a truncated extracellular domain.[46] The truncated extracellular domain creates a new unique targetable peptide sequence.[53; 57] EGFRvIV mutation is less common, seen in approximately 20% of EGFR amplified glioblastomas. EGFRvIV results due to deletion of the carboxyl terminal domain and also exhibits constitutive activation.[48]
Due to high frequency of EGFR alterations and the success in targeting EGFR in other tumors EGFR is an attractive target, however, EGFR inhibitors and an EGFRvIII vaccine have so far had disappointing results in glioblastomas.[58–60] It is reasonable to think that clinical trials targeting this pathway will continue and information regarding EGFR amplification and mutation may be desired. Felsberg et al., van den Bent et al., and Cioca et al. demonstrated that EGFR amplification was retained in recurrent glioblastomas after standard treatment and does not need to be retested. EGFRvIII more commonly shows loss or reduced expression at recurrence and retesting should be considered if targeted therapy is being contemplated.[46; 52; 61] EGFR amplification can be detected by FISH, CGH, or PCR-based assays and EGFR mutations can be detected by PCR or IHC[8; 47; 61; 62] (See Table V).
Synthesis
Testing for EGFR amplification should be performed in cases of IDH-wildtype tumors that are difficult to grade as it may help classify the tumor as a glioblastoma. If a previous EGFR amplification was detected, retesting is not necessary. Repeat EGFR testing may be indicated for patients in which targeted therapy is being considered, particularly therapies targeting specific EGFR mutations.
Question: For adult patients with progressive glioblastoma does whole genome or large panel sequencing provide management or prognostic information beyond that derived from histologic analysis?
Study selection and Characteristics
16 studies were uncovered by the search criteria and ten studies were included in this review. The remaining studies were excluded because they were review articles (4) or were focused on lower grade infiltrating gliomas (2).
Results of individual studies, discussion of study limitations and risk of bias
Currently the only Federal Drug Administration (FDA) approved therapeutic agents for the treatment of progressive glioblastoma are bevacizumab, and in selected patients carmustine-wafers, thus patients are often encouraged to go on clinical trials.[63–65] Glioblastoma is molecularly heterogeneous and appears to be highly mutable with progressive glioblastoma displaying inherent or acquired resistance to treatment.[63; 64; 66]
Whole genome and large panel sequencing have expanded our understanding of the alterations that occur in de novo glioblastoma and progressive glioblastoma. Most studies demonstrate a gain of genetic alterations in recurrent glioblastomas[67] with approximately 17% of recurrent glioblastomas showing hypermutation[68]. Hypermutated tumors with alterations in the retinoblastoma (RB) and mammalian target of rapamycin pathways were noted after temozolomide therapy by Johnson et al.[24] while Wood et al. noted a subset of recurrent glioblastomas demonstrated an increased expression of CHI3L1, TIMP1, and CD44, whose expression has been associated with a more aggressive course[69]. Interestingly in one study 43% of recurrent gliomas showed a loss of 50% or more of the mutations present in the initial tumor, some of which were driver mutations, including TP53, ATRX, SMARCA4, and BRAF[24].
The increase in genetic understanding has also sparked hope that more effective novel or targeted therapies may be available for particular subsets of patients. To this end multiple studies looking at possible targets, alterations in recurrence, and clinical trials involving targeted therapies have been performed or are underway. Most studies were performed using next generation sequencing to asses genomic alterations either using data from whole transcriptome sequencing, whole exome sequencing, or targeted panels. Targeted panels have the advantage of being the most cost effective with the analysis of the data being the most straight forward and having relatively high specificity however the genes included on the targeted panels can vary and requires enrichment of the target regions. Whole exome sequencing requires enrichment of the exons and analysis of the data to ensure optimal processing and sequencing reaction. Whole genome sequencing while giving a comprehensive view of all alterations is the most expensive and requires the most data analysis and interpretation and many of the identified alterations may not be therapeutically targetable or relevant to diagnosis. Additional methods of genomic profiling that can be utilized include chromosomal microarray analysis and DNA methylation. It has become clear that progressive glioblastoma demonstrates an evolution of molecular alterations relative to primary glioblastoma.[24; 67; 69-71] Byron et al. demonstrated that genome wide molecular testing to guide therapy was feasible, and had promising results in 2 patients, in this study of a very limited size.[65] The ability of chosen therapies to cross the blood-brain barrier was taken into account in this study and is an important consideration when designing treatment recommendations. Most targeted therapies, particularly when used as single agent therapy, seem to have limited activity in the setting of progressive glioblastoma.[64] However, given the limited response to therapy in the recurrent setting repeat molecular testing would be of value in patients who are eligible for clinical trials based on a targeted therapy[65; 67; 68](See Table VI).
Synthesis
Primary or repeat whole genome or large panel sequencing should be considered in patients in whose management may be impacted including those who are eligible or interested in targeted therapy based on a particular oncogenic pathway anomaly or pathway member or for assessment of eligibility in clinical trials based on a particular molecular characteristic.
Question: For adult patients with progressive glioblastoma should immune checkpoint biomarker testing be performed to provide management and prognostic information beyond that obtained from histologic analysis?
Study selection and Characteristics
Twenty-seven articles were uncovered during the screening process, however the majority of these represented review articles and in vitro or animal models. Other studies were excluded due to a lack of pathology data (radiology studies), phase 1 studies that did not include pathology or outcomes, and studies focused on pediatric patients. Two additional studies were identified from references of review articles identified in the screening process. Ultimately eight studies were included in the review.
Results of individual studies, discussion of study limitations and risk of bias:
Immune checkpoint inhibitors have shown marked success in the treatment of a variety of solid cancer types by blocking immune checkpoint signaling and allowing a T-cell response against the tumor.[72] Glioblastoma is known to cause host immunosuppression through a variety of mechanisms[63] and glioblastomas show frequent genetic and epigenetic alterations which potentially may produce numerous neoantigens,[72] thus immune checkpoint inhibitors sound like a promising treatment modality for progressive glioblastoma.
Variable PD-L1 staining patterns are seen in glioblastoma ranging from clear membranous staining to diffuse cytoplasmic staining[72; 73] and shows heterogeneity within the tumor.[72] The clinical significance of these differential staining patterns is yet to be elucidated but likely contributes to the wide range of PD-L1 expression in glioblastomas being reported, from 10–88%.[73–76] A large proportion of tumors have been reported to demonstrate the diffuse pattern of staining, up to 88% of primary glioblastomas and 72% of recurrent glioblastomas .[73] While the clear membranous staining pattern is only seen in a subset of primary and progressive glioblastomas, approximately 37% of primary glioblastomas and 11-16.7% of progressive glioblastomas[73; 77], with Berghoff et al. noting a significant decrease in progressive glioblastomas demonstrating clear membranous PD-L1 expression. Heyneckes et al. found a significant decrease in both the PD-L1 mRNA expression and number of PD-L1 positive cells in progressive glioblastomas and that this reduction was more pronounced in patients who received extended temozolomide therapy .[72] The recent CheckMate 143 clinical trial, the first large randomized clinical trial of nivolumab, a PD-1 inhibitor, failed to extend overall survival in the setting of progressive GBM.[78–80] PD-L1 expression was not included as a criteria for inclusion in this study, and in the phase II CheckMate 143 studies while 68% of patients had PD-L1 expression over 1% only 27% of patients had PD-L1 expression greater than 10%. Confounding these values is that the PD-L1 expression was measured on the primary resection, not the recurrent tumor.[78] A phase 1a study of atezolizumab in progressive glioblastoma also demonstrated dismal results with 100% of patients discontinuing therapy due to progressive disease. However, a more recent trial using pembrolizumab in patients with progressive glioblastoma as neoadjuvant therapy prior to re-resection and continuing as adjuvant therapy demonstrated improved overall survival (13.7 months) and progression free survival (3.3 months) when compared to patients receiving pembrolizumab as adjuvant therapy only after re-resection (7.5 months and 2.4 months respectively). Further studies will need to be done to further validate the use and efficacy of pembrolizumab in the neoadjuvant setting as this was a small study, it is also notable that PD-L1 expression was not a criteria and expression was not reported in either cohort.[81]
Another biomarker of interest is loss of mismatch repair (MMR) proteins and in 2017 the FDA approved pembrolizumab (Keytruda) for the treatment of unresectable or metastatic solid tumors harboring mismatch repair deficiency (dMMR) or microsatellite instability-high (MSI-H) regardless of site.[82] MMR enzymes, including MSH2, MSH6, MLH1, and PMS2, are involved in inducing programmed cell death in tumor cells damaged by alkylating agents, including temozolomide.[83] Multiple studies have shown that progressive glioblastoma has an increased prevalence of inactivating mutations of mismatch repair genes, particularly involving MSH6 and there is some evidence that MMR gene alterations are caused by or selected for by temozolomide therapy .[84-86] Indraccolo et al. found that the majority (78.5%) of cases lacking MMR protein expression at recurrence had MGMT promoter methylation at diagnosis.[84] Loss of MMR enzyme expression is also associated with a hypermutant genotype, while this is a small subset of patients (~10%), it is hypothesized that the increased mutagenesis may make these tumors more immunogenic and thus more amenable to immunotherapy[74; 84] (See Table VII). However, the efficacy of pembrolizumab or other immune checkpoint inhibitors has yet to be investigated in MMR deficient progressive glioblastomas has yet to be fully investigated.
Synthesis
If immune checkpoint inhibitors are being considered PD-L1 expression or loss of MMR enzyme activity should be determined but due to the limited benefit demonstrated by immune checkpoint agents in glioblastoma (primary or progressive) standard testing is not currently necessary.
Question: For adult patients with glioblastoma are Bevacizumab biomarkers available and should they be performed in the setting of progressive glioblastoma?
Study selection and Characteristics
Numerous studies (greater than 75) looking at Bevacizumab were identified however most were clinical trial papers, without examination of pathology, studies looking at radiologic biomarkers, or review articles. Eight articles looking at tissue biomarkers were identified and included in this review.
Results of individual studies, discussion of study limitations and risk of bias
Glioblastoma demonstrates marked up-regulation of VEGF-A and displays rapid vascularization.[4; 87] Bevacizumab is a monoclonal antibody against vascular endothelial growth factor (VEGF) that has been approved for treatment of recurrent glioblastoma. However, bevacizumab has not been shown to improve OS and there is concern that glioblastomas treated with bevacizumab are more aggressive and show increased infiltration.[4; 88; 89] A subset of patients do show a favorable clinical response following bevacizumab and biomarkers are being investigated to identify these patients.[4] Choi et al. and Hovinga et al. both found that classical subtype glioblastomas did not respond as well to bevacizumab.[4; 47] Choi et al. and Erdem-Eraslan et al. identified possible biomarkers in predicting response to bevacizumab (COL4A2) and bevacizumab combined with lomustine (FMO4/OSBPL3), however, both of these are retrospective studies on primary tumor specimens and further studies need to be performed to confirm these findings.[4; 90] YKL-40 mRNA expression and plasma levels have been found to be associated with a worse response to carmustine plus bevacizumab or bevacizumab therapy alone, respectively[91; 92] in retrospective studies but no prospective studies have been performed. At this time no established biomarkers are available to predict response to bevacizumab[88; 93; 94] (See table VIII).
Synthesis
Bevacizumab biomarker testing at this time remains experimental, markers which may be indicative of response include YKL-40, COL4A2, and FMO4 expression although confirmatory studies are warranted.