Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Galuppini et al.

Cancer Cell Int (2019) 19:209


https://doi.org/10.1186/s12935-019-0929-4 Cancer Cell International

MINIREVIEW Open Access

Tumor mutation burden:


from comprehensive mutational screening
to the clinic
Francesca Galuppini1†, Carlo Alberto Dal Pozzo1†, Jutta Deckert2, Fotios Loupakis3, Matteo Fassan1*‡ 
and Raffaele Baffa2*‡

Abstract 
The recent advent of immunomodulatory therapies into the clinic has demanded the identification of innovative
predictive biomarkers to identify patients most likely to respond to immunotherapy and support the design of
tailored clinical trials. Current molecular testing for selection of patients with gastrointestinal or pulmonary carcino-
mas relies on the prevalence of PD-L1 expression in tumor as well as immune cells by immunohistochemistry and/
or on the evaluation of the microsatellite status. Tumor Mutational Burden (TMB) has emerged as a promising novel
biomarker in this setting to further aid in patient selection. This has been facilitated by the increasing implementation
of molecular pathology laboratories with comprehensive next generation sequencing (NGS) technologies. However,
the significant overall costs and expertise required for the interpretation of NGS data has limited TMB evaluation in
routine diagnostics, so far. This review focuses on the current use of TMB analysis in the clinical setting in the context
of immune checkpoint inhibitor therapies.
Keywords:  Tumor mutation burden, Immunotherapy, Next generation sequencing, Target therapy

Tumor mutational burden and checkpoint High mutational burden is typical of cancers developed
inhibition as a consequence of exposure to powerful carcinogens,
Cancer has been described as a genomic disease driven such as tobacco smoke and polycyclic aromatic hydrocar-
by an accumulation of both germline derived and somatic bons in lung cancers and bladder cancers, as well as expo-
mutations. Recent advances in molecular techniques sure to mutagens, such as ultraviolet light in melanoma.
have allowed the evaluation of the impact of germline and In recent years, the interest in TMB by physicians and
tumor mutational status on overall cancer risk, patient researchers has increased as tumors with higher TMB
prognosis and treatment response. Tumor mutation fre- can be more responsive to immune checkpoint inhibi-
quency varies widely among cancer types and between tor therapies, which may be due to their increased
tumors of the same histotype. Tumor Mutational Bur- inherent immunogenicity [2]. An effective host anti-
den (TMB) can be analyzed by various methods and tumor immune response requires tumor cell surface
is reported as the total number of sequence variants or antigen recognition followed by priming and activation
mutations per tumor genomic region analyzed [1]. of immune cells that can ultimately mediate tumor cell
killing. However, inhibitory receptor interactions on
immune cells are often hijacked by tumors to dampen
cytotoxic T cell responses against transformed cells
*Correspondence: matteo.fassan@unipd.it; raffaele.baffa@servier.com

Francesca Galuppini and Carlo Alberto Dal Pozzo—Co-first authors
thereby avoiding immune surveillance. For example,

Matteo Fassan and Raffaele Baffa—Co-last authors programmed death ligand 1 (PD-L1) expressed on
1
Department of Medicine, Surgical Pathology Unit, University of Padua, tumors engages the immune checkpoint PD-1 on cyto-
Via Aristide Gabelli, 61, 35121 Padua, Italy
2
Servier Pharmaceuticals, Boston, MA, USA
toxic T lymphocytes to block their action against tumor
Full list of author information is available at the end of the article cells [3]. Likewise, Cytotoxic T lymphocyte antigen 4

© The Author(s) 2019. This article is distributed under the terms of the Creative Commons Attribution 4.0 International License
(http://creat​iveco​mmons​.org/licen​ses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium,
provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license,
and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/
publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
Galuppini et al. Cancer Cell Int (2019) 19:209 Page 2 of 10

(CTLA-4) engagement by B7-1 (CD80)/B7-2 (CD86) Clinical impact of TMB


ligands constitutes another key inhibitory checkpoint The translational significance of TMB assessment is
signal that limits T-cell activation. Immune checkpoint derived from its link to tumor immunogenicity and its
modulating drugs aim to remove these inhibitory sig- subsequent prognostic and predictive values. In fact, sev-
nals to boost the immune response against cancer cells eral recent studies have demonstrated that TMB can be
and relieve innate as well as adaptive immune resist- exploited as a biomarker, especially in order to predict
ance developed by tumors. It has been suggested that patient responses to immune checkpoint modulatory
patients who do not derive benefit from immune check- agents (Table 1).
point inhibitor therapies lack pre-existing anti-tumor The development of drugs against well-described
T-cell responses, in part due to low immunogenicity of PD-1/PD-L1 and CTLA-4/B7 checkpoints interactions
their underlying disease [4–6]. Mutational load, and in have proven effective in unleashing a cytotoxic response
particular, nonsynonymous mutations, in cancer cells against malignant cells and have revolutionized the ther-
may generate novel antigens (termed neoantigens) that apeutic approaches to various solid tumors, including
are not subject to immune tolerance and allow for an those typically marked by strong resistance to traditional
adaptive immune response by the host (Fig.  1). The chemotherapeutics [4]. This is illustrated by the particu-
observation that nonsynonymous mutation burden is lar effectiveness of both nivolumab, and pembrolizumab
associated with efficacy of the anti-PD-1 antibody pem- (targeting PD-1) and ipilimumab (targeting CTLA-4)
brolizumab is consistent with this hypothesis [3, 7]. against a subset of patients with Non-Small Cell Lung
Moreover, several preclinical [8–13] and clinical [14– Cancer (NSCLCs) and melanoma. Despite the significant
17] reports have demonstrated that neoantigen-specific benefit potentially achievable with these new therapies,
effector T cell response lies at the core of recognizing response rates vary widely between cancer types and
and eliminating established tumors. there is a particular need for predictive biomarkers that
It is however important to note that TMB alone does can be applied upfront to identify patients more likely
not represent a direct evidence of tumor immunogenic- to respond to immune checkpoint modulators. To this
ity, due of the complex dynamics that underlie the host end, the predictive value of various features of tumors,
immune response in the context of tumor cells and host immune cells and the tumor microenvironment
their microenvironment. have been further explored including PD-L1 expression

Fig. 1  Schematic diagram of tumor cell with high TMB and its relationship with the immune system. The formation of neoantigens enhanced
immune cell recognition and the effectiveness of immunotherapy
Table 1  Representative recent studies describing the impact of tumor mutation load (TMB) evaluation in the clinical setting
Type of cancer and stage No of investigated patients Test for TMB Cut-off Drug/treatment Results References

SCLC 211 (133 Nivo, 78 Nivo + Ipi) WES ≥ 248 total mut Nivolumab, Nivolumab + Ipili- Improved ORR, 1y PFS and 1y OS for [21]
mumab TMB high vs. TMB medium and
low patients
Galuppini et al. Cancer Cell Int

NSCLC 34 (16 discovery, 18 validation) WES ≥ 178 total mut Pembrolizumab Higher TMB was associated with [20]
improved objective response,
durable clinical benefit and PFS
NSCLC 312 (158 Nivo, 154 Chemo) WES ≥ 243 total mut Nivolumab vs chemotherapy High TMB associated with increased [22]
ORR and PFS, but not OS
NSCLC 240 NGS (49 also with WES) Anti-PD-(L)1 monotherapy or in Elevated TMB improved likelihood [30]
(2019) 19:209

combination with anti-CTL-4 of benefit to ICIs


Targeted NGS accurately estimates
TMB and correlates with WES
results
Stage IV or recurrent NSCLC 299, 139 (Nivo + Ipi), 160 (chemo- NGS Nivolumab plus ipilimumab, vs. PFS was longer with first line [40]
therapy) chemotherapy nivolumab plus ipilimumab than
with chemotherapy and a high
TMB, irrespective of PD-L1 expres-
sion level
Metastatic melanoma 65 (32 + 33) NGS Nivolumab or pembrolizumab or Response rate, PFS and OS were [6] l
atezolimumab superior in high mutation load
group
Metastatic melanoma 64 (25 discovery +39 validation) WES > 100 total mut Ipilimumab or tremelimumab Mutational load is associated with [18]
the degree of clinical benefit
CRC​ 6004 Comprehensive – TMB classifies MSI tumors as TMB- [27]
Genomic Profiling high and identifies nearly 3% of
(CGP) CRC as MSS/TMB-high
CLL 91 WES allo-HSCT Clinically evident durable remission [15]
in patients with neoantigen
peptides
26 cancer types 11,348 NGS and MSI-NGS – MSI offers distinct data for treat- [28]
ment decision regarding immune
checkpoint inhibitors, in addition
to TMB and PD-L1
12 cancer types 86 MSI-NGS Pembrolizumab Large proportion of mutant neoan- [29]
tigens in MSI cancers make them
sensitive to immune checkpoint
blockade, regardless the cancer’s
tissue of origin
NSCLC Non-small cell lung cancer, WES whole-exome sequencing, PFS progression-free survival, OS overall survival, allo-HSCT allo-hematopoietic stem cell transplantation, TILs tumor-infiltrating lymphocytes, MSI
microsatellite instability, ICIs immune checkpoint inhibitors
Page 3 of 10
Galuppini et al. Cancer Cell Int (2019) 19:209 Page 4 of 10

in both tumor and immune cells, selected single gene of DNA repair pathway mutations, but not with patient
mutational status, peripheral-blood lymphocyte count, HLA haplotype. Of note, the authors provided a TMB
tumor infiltrating lymphocyte count, markers of T-cell threshold (based on nonsynonymous mutation burden)
activation and evaluation of inflammatory cytokines suitable to identify candidate patients in clinical prac-
[18]. Nevertheless, none of these have been unambigu- tice: in the validation cohort a cut-off of 178 mutations
ously associated with patient outcome endpoints includ- per tumor provide sensitivity of about 85% and speci-
ing Overall Survival (OS), Progression Free Survival ficity of about 75% in differentiating responders from
(PFS), and Objective Response Rate (ORR) across mul- non-responders.
tiple tumor types. To date, microsatellite instability-high Building on the positive predictive value of high TMB
(MSI-H) status, mismatch repair deficiency and PD-L1 for the response to anti-PD1/PD-L1 and anti-CTLA-4,
expression constitute the only predictive biomarkers suc- many subsequent analyses focused on identification of
cessfully used for patient selection in select malignancies. standardized cut-offs suitable to harmonize the results of
Since TMB assessment is not exempt from economical various clinical trials and the relationships of TMB with
and technical issues, it is important to establish a list of other biomarkers, in particular PD-L1 expression.
malignancies that are more likely to be highly mutated A crucial analysis by Hellman and co-workers [21]
and, thus, to be priority candidates for this analysis. on > 200 SCLC patient samples from the CheckMate
Here the etiology of the diverse neoplastic patholo- 032 trial compared outcomes for patients treated with
gies plays a critical role. In principle, TMB is likely to be nivolumab or a combination of nivolumab and ipili-
high especially in two categories of tumors: (i) those that mumab. Patients were grouped into tertiles based on
arise from the exposure to powerful carcinogenic and TMB analysis and this analysis revealed improved ORR
mutagenic agents (e.g. tobacco smoke and UV-A) and (ii) and favorable 1-year PFS and OS outcomes for TMB-
those caused by germline mutations in genes encoding high (≥ 248 mutations) patients treated with nivolumab
for proteins involved in DNA repair and replication. or a combination of nivolumab and ipilimumab com-
pared to patients with medium or low TMB. In contrast,
TMB in lung cancer PD-L1 expression status was not predictive of response
The predictive role of TMB in pulmonary metastatic and no association was found between PD-L1 expression
tumors constitutes a remarkable paradigm [19]. Indeed, and TMB.
immune checkpoint inhibitor therapies have proven In the Phase 3 CheckMate 026 trial in first-line PD-L1
effective against pulmonary malignancies, whose etiology positive NSCLC patients, nivolumab treatment was not
is in most cases linked to exposure to the “carcinogenic associated with improved ORR or prolonged PFS versus
cocktail” contained in cigarette smoking [19]. In fact, chemotherapy. However, an exploratory analysis of 312
both TMB rate and effectiveness of immune checkpoint patients evaluated the effect of TMB on outcomes, with
inhibitors are higher in neoplasms developed by smokers patients divided into tertiles based on TMB status. ORR
in comparison to the ones developed by never-smokers. and PFS were improved in the TMB-high group (≥ 243
This is likely a result of the “signature of smoking” [20] mutations) treated with nivolumab as compared to
which is characterized by a transversion-high mutational chemotherapy, while OS was similar [22]. Again, PD-L1
profile that results in an increased number of non-syn- expression was not predictive of response, even for a sub-
onymous mutations and, ultimately, in a greater neoan- set with PD-L1 ≥ 50%, and did not correlate with TMB.
tigens load. Furthermore, efficacy of immunomodulatory Similarly, in a subgroup analysis of the Phase 3 Check-
therapies in Small Cell Lung Cancer (SCLCs) appears to Mate 227 trial on ~ 300 metastatic NSCLC patients with
be lower than in NSCLCs, while SCLC is also marked TMB of > 10 mutations/Mb, combination treatment with
by a lower number of mutations per megabase than Nivolumab and Ipilimumab resulted in prolonged PFS
NSCLCs. compared to chemotherapy treatment [40]. Importantly,
A crucial study published in 2015 by Rizvi et  al. [20] in this study TMB was an independent positive predic-
demonstrated that higher nonsynonymous mutation tive biomarker, irrespective of tumor PD-L1 expression
burden in tumors was associated with improved ORR, level. The value of TMB assessment was also confirmed
durable clinical benefit (DCB), and prolonged PFS in a in a multivariate analysis including sex, tumor histo-
retrospective analysis of two cohorts of NSCLC patients logic type and ECOG performance-status. Therefore, the
treated with the anti-PD-1 pembrolizumab. Furthermore, authors suggested that combined therapy with nivolumab
this analysis suggested that the efficacy of immune check- plus ipilimumab may represent an effective first-line
point inhibition by pembrolizumab in the treatment treatment regimen for patients with advanced NSCLCs
of NSCLC also correlated with the molecular smoking marked by high TMB, irrespective of PD-L1 expression
signature, higher neoantigen burden, and the presence level and other investigated clinical and pathological
Galuppini et al. Cancer Cell Int (2019) 19:209 Page 5 of 10

variables. Unfortunately, final analysis of this trial popu- of TMB, which could represent a practical strategy to
lation is still pending, making it uncertain if TMB status overcome the limits of a single threshold value in terms
will achieve regulatory approval as a predictive biomarker of sensitivity and specificity.
for this treatment regimen in metastatic NSCLC.
In another study, 240 NSCLC patients treated with TMB in other cancers
anti-PD-1 or anti-PD-L1 based therapy were profiled for A study by Powles et  al. [26] investigated outcomes of
TMB by targeted NGS. TMB was significantly associated metastatic platinum-refractory urothelial carcinoma
with improved patient outcomes with the increased odds patients treated with atezolizumab versus chemotherapy
of disease control with increasing thresholds [30]. Impor- in the IMvigor211 trial. Here, high PD-L1 expression
tantly, comparison of NGS with WES analysis showed was not a predictive biomarker. However, in an explora-
high correlation between these methods. tory analysis in > 500 samples, high TMB correlated with
increased OS in patients treated with atezolizumab, sup-
TMB in melanoma porting the role of TMB as an alternate biomarker.
The etiology of melanoma is typically related to the muta- In addition, CRC presents a perfect opportunity to
genic effects of UV exposure. Comprehensive sequenc- study the role of DNA-repair deficiency in the develop-
ing data (i.e. 507 whole genome and 6535 whole exome ment of high TMB. In an elegant study by Fabrizio et al.
sequences) from the International Cancer Genome Con- [27] TMB assessment has been connected to microsatel-
sortium (ICGC) pinpointed malignant melanoma as lite status and DNA-repair genes mutational status in a
the tumor with the highest mutation prevalence among cohort of patients with metastatic CRCs. Here almost
27 different histotypes [23]. The genotoxic effects of all (301/302, 99.7%) MSI-high patients were classified as
the UV-A are comparable to those of smoking. Studies TMB-high and their tumors were marked by mutations
of Weber et  al. (including patients with melanoma who in DNA-repair genes such as MSH2, MSH6 and MLH1.
have progressed after being treated with ipilimumab and This suggests that MSI-high status could serve as a surro-
BRAF inhibitors) [24] and Robert et  al. [25] (including gate biomarker of high TMB in metastatic CRCs in order
metastatic melanoma patients, negative for BRAF muta- to predict response to immune checkpoints modulation
tion, not treated previously) reported that checkpoint therapy. However, MSI-high status alone may not capture
inhibitors have been particularly successful in melanoma, all metastatic CRCs potentially sensitive to anti-PD1/
with the highest response rates to single-agent PD-1/ PD-L1 drugs. In fact, in the same study MSI-high tumors
PD-L1 inhibition. In this indication, the connection accounted for only for 97% of all TMB-high metastatic
between TMB and tumor immunogenicity is again high- CRCs. Thus, an estimated 3% of high-TMB metastatic
lighted by the fact that non-cutaneous melanomas have CRC tumors are microsatellite stable (MSS). Due to the
far fewer mutations than those of cutaneous origin, and high prevalence and mortality related to metastatic CRC
show are less responsive to immunotherapy. (about 50,000 deaths/year), the correct identification of
In an early report from 2014, Snyder et  al. [18] found TMB-high/MSS malignancies might lead to a substan-
that high mutational load correlates with a sustained tial therapeutic benefit for an additional 1500 patients/
clinical benefit from CTLA-4 blockade in patients suffer- year. These considerations underline once again the criti-
ing from metastatic melanomas. Nonetheless, high TMB cal predictive value of comprehensive TMB assessment,
alone is clearly not sufficient for clinical benefit, as not all which is cannot be easily replaced by other surrogate
tumors with a high mutational burden responded to ther- biomarkers.
apy. This study further identifies the existence of specific Therefore, the importance of TMB, microsatellite and
tumoral neoantigens that create a neoepitope signature PD-L1 status and the interplay between them differs sig-
as critical for the response to anti-CTLA-4 and overall nificantly between cancer types which may be context
high mutational load increased the probability of such a and mechanism dependent. Thus further investigations
signature being present in melanoma patients. are needed to better understand their respective transla-
Another retrospective study reported by Johnson et al. tional value alone and in combination [28]. In the future,
[6] evaluated the predictive role of TMB in two cohorts this will ideally enable more precise patient stratification
of patients with metastatic melanoma treated with anti- based on assessment of multiple biomarkers, preferably
PD-1/PD-L1 antibodies. In both cohorts ORR, PFS and from the same patient biopsy.
overall survival (OS) was superior in the high, compared
to intermediate and low mutation load groups. This is the Testing TMB: the technical approach
first demonstration of TMB as an independent prognos- The gold standard and most widely used method to assess
tic value for multiple immune checkpoints-modulators. TMB for research purposes is whole exome sequencing
Moreover, the authors introduced a stratified evaluation (WES). Indeed, many of the studies cited above have used
Galuppini et al. Cancer Cell Int (2019) 19:209 Page 6 of 10

WES as the basis for the retrospective TMB evaluation. actionable genomic targets in the gene panel [32, 33]
However, this technique is expensive and requires exces- may help to further refine the molecular determinants of
sively lengthy turnaround times making it unsuitable for response to immunotherapy, going beyond the “simple”
large scale and routine clinical applications [2]. To over- assessment of TMB.
come this problem, two alternative approaches have been
proposed: (i) the development of “surrogate biomarkers” TMB assessment: open issues
that could be easily evaluated; (ii) the indirect determi- While it is now apparent that TMB is a valuable bio-
nation of TMB by evaluation of the mutational status marker to predict the response to PD-1/PD-L1 and
of a defined gene panel by Next Generation Sequencing CTLA4/B7-1 axes inhibitors, there are still several prob-
(NGS). lems that impede the routine adoption of this predictive
The intrinsic shortcoming of the first proposal is that approach. These barriers reside in the analysis itself as
any surrogate biomarker would represent an approxima- well as the pre- and post-analytic phases.
tion for TMB, which in itself is a representation of tumor Widespread implementation of TMB testing in the
immunogenicity. Any indirect TMB assessment will clinic is still challenging. Sample size, sample quality and
likely introduce an even greater margin for error. Overall, the resulting DNA yields are rate-limiting factors from
biomarkers suitable for this purpose include MSI status the patient biopsy perspective. Cost associated with test-
[27, 29] (see above), chromosomal structural analyses ing, including the need for specialized equipment and
[19] and mutational analyses of selected genes. The lat- highly-trained personnel, can further limit the imple-
ter approach might be useful in specific tumor subtypes, mentation of the TMB analysis in a routine setting. In
despite the existence of the abovementioned intrinsic addition, varying testing platforms, different bioinfor-
issues. For example, metastatic melanomas bearing muta- matic pipelines, and non-standardized cut-off definitions
tions in LRP1B gene have a significantly higher muta- hinder comparison between data sets from different sites
tional load as compared with LRP1B wild type tumors; and studies described in the literature. It is apparent that
furthermore, mutational load correlates with the number standardization of TMB evaluation is needed in order to
of LRP1B mutations per tumor [6]. This is likely due to ensure reliability, reproducibility and clinical utility [34].
the size and chromosomal location of LRP1B. Indeed, Size of the NGS gene panel is critical for accurate anal-
this is a large putative tumor suppressor gene, located in ysis. It has been shown that as NGS panels for TMB anal-
a common fragile chromosomal site. Due to these unique ysis become smaller in size, the uncertainty associated
structural characteristics, LRP1B mutational status is with TMB estimation increases rapidly. Specifically, the
likely a good approximation of total exonal mutational coefficient of variance increases rapidly when the size of
load. Other genes whose mutational status might be the targeted panels is less than 1 Mb [35]. Therefore, the
associated with high TMB are KRAS in NSCLCs (due to minimum size of the panel for determining the TMB of
its link to the smoking signature), MSH2, MSH6, MLH1 more than 300 genes or 1 Mb has been proposed.
in CRCs (due to the relationship between MSI and high Scoring of detected alterations has not been standard-
TMB) [27, 28] and finally NF1, BRAF, and NRAS in meta- ized. Although the higher occurrence of synonymous
static melanomas [6]. variants may indicate a mutational process that also
In contrast, NGS appears to be a more powerful tool results in nonsynonymous changes, synonymous and
to translate TMB assessment in clinical practice. Many germline variants are commonly discarded in the calcu-
studies [2, 4, 19, 28, 30] have demonstrated that TMB lation of TMB, as it is assumed that these variants are
evaluation by comprehensive genomic profiling on NGS unlikely directly involved in neoantigen generation. In
diagnostic platforms correlated well with gold-standard the setting of tumor-only sequencing, germline false pos-
WES evaluation. This approach can provide several nota- itive variants may be filtered out by using large and avail-
ble advantages: (i) NGS is available in many academic able germline variant data sets. The use of these germline
centers and it has already become commonplace in clini- databases is a critical step in this process, and it is neces-
cal oncology [2]; (ii) NGS is less expensive than WES; sary to use germline databases with a sufficiently broad
(iii) NGS analysis shortens the assay turnaround times representation of all populations. The variability of the
to < 10  days; which is the recommended timeframe for TMB among different studies is due also to different scor-
clinical decision making on the basis of molecular char- ing of alterations, as some studies consider all alterations
acterization of neoplasms [31]; (iv) in addition to TMB, including the copy number alterations [30], while others
NGS can also provide information on other critical exclude copy number changes [36], variants included in
prognostic and predictive factors (e.g. EGFR, KRAS, and COSMIC or alterations that are likely to be or are known
BRAF in NSCLCs or KRAS, NRAS and BRAF in CRCs). to be bona fide oncogenic drivers and germline polymor-
In addition, the simultaneous assessment of multiple phisms [37] in order to avoid bias.
Galuppini et al. Cancer Cell Int (2019) 19:209 Page 7 of 10

In clinical practice, the testing of mutational burden immunogenicity. Nevertheless, immunogenicity is over-
should be performed early during the decision-making all a multifactorial feature, which is only partially due to
process in order to select the most appropriate first- the mutational load. TMB alone cannot accurately assess
line treatment. Thus, a sufficient quantity and quality of the dynamic immune status of both tumor and tumor
tumor samples is required [21, 38]. Nevertheless, biopsy microenvironment. Therefore, it is rational to assume
collection can be difficult as patients who are candidates that the comprehensive and integrated evaluation of
for immunotherapy often suffer from metastatic disease genomic (TMB, single gene mutations, MSI etc.) [27],
by definition and often present with poor performance immunohistochemical (e.g. PD-L1 expression) [19, 21]
status (PS). As a consequence, not all patients will have and histological (e.g. tumor grading and tumor infiltrat-
sufficient tumor tissue available or will be able to safely ing lymphocytes—TILs) [19] variables can significantly
undergo a biopsy. Therefore, less invasive sampling meth- refine the selection of patients candidate for treatment
ods are warranted. In this perspective, the study of circu- with immune-checkpoints-modulators.
lating tumor DNA could provide a non-invasive method Another major obstacle to make TMB assessment clini-
for assessing TMB, but further research is needed to cally meaningful is the lack of a widely approved cut-off
establish the performances of this approach in clinical score (and an associated NGS panel) which must be set
settings. to successfully establish TMB as a predictive biomarker.
While the ongoing debate about the best method for To date, no agreement has been reached between the
TMB evaluation favors NSG over WES and evaluation various authors. Some authors prefer a single threshold
of surrogate biomarkers for technical and practical rea- value to select patients for the administration of immune-
sons, standards have not been established for gene panels checkpoint-modulators. For example, in the cohorts
used in NSG approaches. In addition, implementation of studied by Rizvi et al. using WES [2] a load of at least 178
an organizational framework for the next generation sur- mutations per exome provided a sensitivity of 86% and a
gical pathology laboratories is crucial, in order to maxi- specificity of 75% in identifying patients who can benefit
mize the impact of TMB assessment on clinical decision from immunotherapy. Studies applying NGS an select
making. Current guidelines recommend a timeframe of 3 gene panels with approximately 200–300 genes have
workdays from a request for testing to receipt by a refer- either categorized TMB simply as high (at or above the
ence laboratory and 10 workdays for availability of testing median) or low (less than the median) without specifying
results [32, 33, 39]. According to observation of Vander- a discreet cut-off value [26, 39] or have set a single cut-off
Laan et  al. [31] in their institution (Beth Israel Deacon- [27, 33]. In many studies, malignancies have been further
ess Medical Center Boston, MA) these objectives were stratified in several classes according to their mutational
reached in only 50% of cases when a comprehensive NGS load (e.g. low/intermediate/high TMB) [6, 19]. This
assay was performed. These findings demonstrate that approach has the virtue of allowing a greater level of
a turnaround time of 10 workdays is indeed technically therapeutic personalization in the future although this is
feasible with the current available technologies, since half yet untested. For example, patients with high TMB may
of the samples has been successfully analyzed within that be more likely to respond to a single-drug immunomodu-
period. Yet, they also highlight that reliable testing work- latory treatment, while patients bearing an intermediated
streams have not been established and while the ability mutational load could benefit from more aggressive but
to comply with current guidelines might differ between potentially more toxic combination regimens (e.g. anti-
various centers, multidisciplinary efforts are needed in PD-L1 combined with anti CTLA4 antibodies).
order to continuously improve turnaround times. These Finally, even if we were able to efficiently determinate
should include efficient workflows for automated order- and interpret TMB predictive value, there are still impor-
ing of molecular profiling, investment in infrastructure to tant open issues in the choice of best immunomodula-
expedite data delivery, and improved software to stream- tory treatment for each patient. Unaddressed questions
line somatic variant analysis [31]. concern the role of single agent treatments compared
Once TMB data have been successfully collected, the with combination treatments, the potential association
main bottleneck is their interpretations. The main prob- between immunotherapy and chemotherapy and the
lems derive from (i) the intrinsic complexity of biologi- preferred sequencing of therapies. Furthermore, there
cal meaning of TMB; (ii) the lack of an unambiguous key is an open question of how TMB analysis impacts the
to understand the data, and (iii) the multiple therapeutic risk–benefit balance in patients receiving immune check-
protocols put in place for highly-mutated malignancies. point inhibitors. The study by Hellmann [40] evaluated
With regard to the first issue, it has been already efficacy outcomes for several treatment regimens (co-
mentioned that the translational value of TMB assess- primary endpoint PFS with Nivolumab + Ipilimumab vs
ment lies in its assumed relationship with tumor Chemotherapy in patients with TMB ≥ 10 mut/Mb, and
Galuppini et al. Cancer Cell Int (2019) 19:209 Page 8 of 10

as secondary endpoint PFS with Nivolumab vs Chemo- of PD-L1 expression [40]. Applying our knowledge of
therapy in patients with TMB ≥ 13 mut/Mb and ≥ 1% tumor-subtype etiology may improve the predictive value
PD-L1 expression). Treatment-related adverse events of TMB assessment by identifying the diseases that are a
resulting in discontinuation and serious adverse event priori more likely to bear high mutational load.
rates were higher for the nivolumab + ipilimumab combi- Based on the literature data, wide spread application
nation than chemotherapy in all treated patients and the of TMB analysis in clinical practice is still premature and
subgroup with TMB ≥ 10 mut/Mb. Yet, this combination hurdles remain for routine implementation. However,
treatment also resulted in improved PFS in the TMB- emerging data from large randomized trials currently
high population. ongoing may provide more definite criteria for its inter-
pretation and demonstrate its clinical feasibility.
Conclusions Acknowledgements
This review of current available literature suggests that Not applicable.
TMB is a robust predictive biomarker to select patients Authors’ contributions
for immune checkpoint immunotherapy that could be All authors of this research paper participated directly in the planning and
readily applied in many metastatic cancer settings. This execution of the study, and in the analysis of the results. All the authors
actively participated in the writing of the article. All authors read and
assessment promises to be particularly useful for malig- approved the final manuscript.
nancies developed as a consequence of the exposure
to powerful mutagens and carcinogens (e.g. NSCLCs, Funding
Not applicable.
SCLCs and melanomas) or marked by frequent muta-
tions in genes involved in DNA repair and replication Availability of data and materials
(e.g. MSI-high CRCs). In these indications TMB should Not applicable.
be considered as a reliable tissue-based biomarker for Ethics approval and consent to participate
immune checkpoint blockade. Several comprehensive Not applicable.
studies performed in recent years have provided evi-
Consent for publication
dence for a link between TMB and patient outcomes in Not applicable.
NSCLCs and melanomas, which are among the most
highly mutated human tumors. Subset analysi of TMB- Competing interests
Jutta Deckert and Raffaele Baffa are employees of Servier Pharmaceuticals. The
high patients in other malignancies have supported the other authors have no competing interests to declare. All the authors declare
same observation. no competing interests regarding the publication of this article.
TMB has clearly emerged as a robust predictive bio-
Author details
marker, yet there is still debate about its value by itself 1
 Department of Medicine, Surgical Pathology Unit, University of Padua, Via
rather than in combination with other variables, such as Aristide Gabelli, 61, 35121 Padua, Italy. 2 Servier Pharmaceuticals, Boston, MA,
PD-L1 expression, TILs and critical/driver single-gene USA. 3 Veneto Institute of Oncology, IOV-IRCCS, Oncology Unit 1, Padua, Italy.
mutations. Furthermore, there are remaining logisti- Received: 19 February 2019 Accepted: 4 August 2019
cal issues that hamper implementing TMB evaluation in
daily clinical practice. These include difficulties in obtain-
ing sufficient biopsies from often critically ill patients as
well as lack of a standard analysis method that is eco- References
nomically viable and combines robust performance 1. Yarchoan M, Hopkins A, Jaffee EM. Tumor mutational burden and
with an acceptable turnaround time. In this respect, the response rate to PD-1 inhibition. N Engl J Med. 2017;377:2500–1.
2. Steuer CE, Ramalingam SS. Tumor mutation burden: leading immuno-
use of NGS on selected-gene panels emerges as the best therapy to the era of precision medicine? J Clin Oncol. 2018;36:631–2.
tool currently available to more widely implement TMB 3. Iwai Y, Hamanishi J, Chamoto K, Honjo T. Cancer immunotherapies target-
assessment in clinical practice. Nevertheless, large-sized ing the PD-1 signaling pathway. J Biomed Sci. 2017;24:26.
4. Boumber Y. Tumor mutational burden (TMB) as a biomarker of
randomized trials are warranted to provide and confirm response to immunotherapy in small cell lung cancer. J Thorac Dis.
standardized cut-offs required to confirm TMB assess- 2018;10:4689–93.
ment as a meaningful predictor of patient outcomes. 5. Goto Y. Tumor mutation burden: is it ready for the clinic? J Clin Oncol.
2018. https​://doi.org/10.1200/JCO.2018.79.3398.
Additional characterization of tumor biology such as 6. Johnson DB, Frampton GM, Rioth MJ, Yusko E, Xu Y, Guo X, Ennis RC,
single-gene mutational status, MSI status and presence of Fabrizio D, Chalmers ZR, Greenbowe J, Ali SM, Balasubramanian S, Sun
TILs should not be considered as a substitute for TMB JX, He Y, Frederick DT, Puzanov I, Balko JM, Cates JM, Ross JS, Sanders C,
Robins H, Shyr Y, Miller VA, Stephens PJ, Sullivan RJ, Sosman JA, Lovly CM.
assessment, but can rather complement it to better pre- Targeted next generation sequencing identifies markers of response to
dict individual patient response to immunotherapy. This PD-1 blockade. Cancer Immunol Res. 2016;4:959–67.
is underscored by the observation that in NSCLC TMB 7. Garon EB, Rizvi NA, Hui R, Leighl N, Balmanoukian AS, Eder JP, Patnaik
A, Aggarwal C, Gubens M, Horn L, Carcereny E, Ahn MJ, Felip E, Lee JS,
is an independent predictive biomarker, irrespective
Galuppini et al. Cancer Cell Int (2019) 19:209 Page 9 of 10

Hellmann MD, Hamid O, Goldman JW, Soria JC, Dolled-Filhart M, Rutledge 21. Hellmann MD, Callahan MK, Awad MM, Calvo E, Ascierto PA, Atmaca
RZ, Zhang J, Lunceford JK, Rangwala R, Lubiniecki GM, Roach C, Eman- A, Rizvi NA, Hirsch FR, Selvaggi G, Szustakowski JD, Sasson A, Golhar R,
cipator K, Gandhi L, KEYNOTE-001 Investigators. Pembrolizumab for the Vitazka P, Chang H, Geese WJ, Antonia SJ. Tumor mutational burden and
treatment of non-small-cell lung cancer. N Engl J Med. 2015;372:2018–28. efficacy of nivolumab monotherapy and in combination with ipilimumab
8. Matsushita H, Vesely MD, Koboldt DC, Rickert CG, Uppaluri R, Magrini VJ, in small-cell lung cancer. Cancer Cell. 2018;33(853):861.e4.
Arthur CD, White JM, Chen YS, Shea LK, Hundal J, Wendl MC, Demeter 22. Carbone DP, Reck M, Paz-Ares L, Creelan B, Horn L, Steins M, Felip E, van
R, Wylie T, Allison JP, Smyth MJ, Old LJ, Mardis ER, Schreiber RD. Cancer den Heuvel MM, Ciuleanu TE, Badin F, Ready N, Hiltermann TJN, Nair
exome analysis reveals a T-cell-dependent mechanism of cancer immu- S, Juergens R, Peters S, Minenza E, Wrangle JM, Rodriguez-Abreu D,
noediting. Nature. 2012;482:400–4. Borghaei H, Blumenschein GR Jr, Villaruz LC, Havel L, Krejci J, Corral Jaime
9. Castle JC, Kreiter S, Diekmann J, Lower M, van de Roemer N, de Graaf J, J, Chang H, Geese WJ, Bhagavatheeswaran P, Chen AC, Socinski MA,
Selmi A, Diken M, Boegel S, Paret C, Koslowski M, Kuhn AN, Britten CM, CheckMate 026 Investigators. First-line nivolumab in stage IV or recurrent
Huber C, Tureci O, Sahin U. Exploiting the mutanome for tumor vaccina- non-small-cell lung cancer. N Engl J Med. 2017;376:2415–26.
tion. Cancer Res. 2012;72:1081–91. https​://doi.org/10.1158/0008-5472. 23. Alexandrov LB, Nik-Zainal S, Wedge DC, Aparicio SA, Behjati S, Biankin AV,
CAN-11-3722 Bignell GR, Bolli N, Borg A, Borresen-Dale AL, Boyault S, Burkhardt B, Butler
10. Gubin MM, Zhang X, Schuster H, Caron E, Ward JP, Noguchi T, Ivanova Y, AP, Caldas C, Davies HR, Desmedt C, Eils R, Eyfjord JE, Foekens JA, Greaves
Hundal J, Arthur CD, Krebber WJ, Mulder GE, Toebes M, Vesely MD, Lam M, Hosoda F, Hutter B, Ilicic T, Imbeaud S, Imielinski M, Jager N, Jones DT,
SS, Korman AJ, Allison JP, Freeman GJ, Sharpe AH, Pearce EL, Schumacher Jones D, Knappskog S, Kool M, Lakhani SR, Lopez-Otin C, Martin S, Mun-
TN, Aebersold R, Rammensee HG, Melief CJ, Mardis ER, Gillanders WE, shi NC, Nakamura H, Northcott PA, Pajic M, Papaemmanuil E, Paradiso A,
Artyomov MN, Schreiber RD. Checkpoint blockade cancer immunother- Pearson JV, Puente XS, Raine K, Ramakrishna M, Richardson AL, Richter
apy targets tumour-specific mutant antigens. Nature. 2014;515:577–81. J, Rosenstiel P, Schlesner M, Schumacher TN, Span PN, Teague JW, Totoki
11. Schumacher T, Bunse L, Pusch S, Sahm F, Wiestler B, Quandt J, Menn O, Y, Tutt AN, Valdes-Mas R, van Buuren MM, van ‘t Veer L, Vincent-Salomon
Osswald M, Oezen I, Ott M, Keil M, Balss J, Rauschenbach K, Grabowska A, Waddell N, Yates LR, Australian Pancreatic Cancer Genome Initiative,
AK, Vogler I, Diekmann J, Trautwein N, Eichmuller SB, Okun J, Stevanovic S, ICGC Breast Cancer Consortium, ICGC MMML-Seq Consortium, ICGC
Riemer AB, Sahin U, Friese MA, Beckhove P, von Deimling A, Wick W, Plat- PedBrain, Zucman-Rossi J, Futreal PA, McDermott U, Lichter P, Meyerson
ten M. A vaccine targeting mutant IDH1 induces antitumour immunity. M, Grimmond SM, Siebert R, Campo E, Shibata T, Pfister SM, Campbell PJ,
Nature. 2014;512:324–7. Stratton MR. Signatures of mutational processes in human cancer. Nature.
12. Duan F, Duitama J, Al Seesi S, Ayres CM, Corcelli SA, Pawashe AP, Blan- 2013;500:415–21.
chard T, McMahon D, Sidney J, Sette A, Baker BM, Mandoiu II, Srivastava 24. Weber JS, D’Angelo SP, Minor D, Hodi FS, Gutzmer R, Neyns B, Hoeller C,
PK. Genomic and bioinformatic profiling of mutational neoepitopes Khushalani NI, Miller WH Jr, Lao CD, Linette GP, Thomas L, Lorigan P, Gross-
reveals new rules to predict anticancer immunogenicity. J Exp Med. mann KF, Hassel JC, Maio M, Sznol M, Ascierto PA, Mohr P, Chmielowski
2014;211:2231–48. B, Bryce A, Svane IM, Grob JJ, Krackhardt AM, Horak C, Lambert A, Yang
13. Yadav M, Jhunjhunwala S, Phung QT, Lupardus P, Tanguay J, Bumbaca S, AS, Larkin J. Nivolumab versus chemotherapy in patients with advanced
Franci C, Cheung TK, Fritsche J, Weinschenk T, Modrusan Z, Mellman I, Lill melanoma who progressed after anti-CTLA-4 treatment (CheckMate
JR, Delamarre L. Predicting immunogenic tumour mutations by combin- 037): a randomised, controlled, open-label, phase 3 trial. Lancet Oncol.
ing mass spectrometry and exome sequencing. Nature. 2014;515:572–6. 2015;16:375–84.
14. van Rooij N, van Buuren MM, Philips D, Velds A, Toebes M, Heemskerk B, 25. Robert C, Long GV, Brady B, Dutriaux C, Maio M, Mortier L, Hassel JC,
van Dijk LJ, Behjati S, Hilkmann H, El Atmioui D, Nieuwland M, Stratton Rutkowski P, McNeil C, Kalinka-Warzocha E, Savage KJ, Hernberg MM,
MR, Kerkhoven RM, Kesmir C, Haanen JB, Kvistborg P, Schumacher TN. Lebbe C, Charles J, Mihalcioiu C, Chiarion-Sileni V, Mauch C, Cognetti
Tumor exome analysis reveals neoantigen-specific T-cell reactivity in an F, Arance A, Schmidt H, Schadendorf D, Gogas H, Lundgren-Eriksson L,
ipilimumab-responsive melanoma. J Clin Oncol. 2013;31:e439–42. Horak C, Sharkey B, Waxman IM, Atkinson V, Ascierto PA. Nivolumab in
15. Rajasagi M, Shukla SA, Fritsch EF, Keskin DB, DeLuca D, Carmona E, Zhang previously untreated melanoma without BRAF mutation. N Engl J Med.
W, Sougnez C, Cibulskis K, Sidney J, Stevenson K, Ritz J, Neuberg D, Brusic 2015;372:320–30.
V, Gabriel S, Lander ES, Getz G, Hacohen N, Wu CJ. Systematic identifica- 26. Powles T, Duran I, van der Heijden MS, Loriot Y, Vogelzang NJ, De Giorgi U,
tion of personal tumor-specific neoantigens in chronic lymphocytic Oudard S, Retz MM, Castellano D, Bamias A, Flechon A, Gravis G, Hussain
leukemia. Blood. 2014;124:453–62. S, Takano T, Leng N, Kadel EE 3rd, Banchereau R, Hegde PS, Mariathasan
16. Robbins PF, Lu YC, El-Gamil M, Li YF, Gross C, Gartner J, Lin JC, Teer JK, S, Cui N, Shen X, Derleth CL, Green MC, Ravaud A. Atezolizumab versus
Cliften P, Tycksen E, Samuels Y, Rosenberg SA. Mining exomic sequencing chemotherapy in patients with platinum-treated locally advanced or
data to identify mutated antigens recognized by adoptively transferred metastatic urothelial carcinoma (IMvigor211): a multicentre, open-label,
tumor-reactive T cells. Nat Med. 2013;19:747–52. phase 3 randomised controlled trial. Lancet. 2018;391:748–57.
17. Linnemann C, van Buuren MM, Bies L, Verdegaal EM, Schotte R, Calis JJ, 27. Fabrizio DA, George TJ Jr, Dunne RF, Frampton G, Sun J, Gowen K, Ken-
Behjati S, Velds A, Hilkmann H, Atmioui DE, Visser M, Stratton MR, Haanen nedy M, Greenbowe J, Schrock AB, Hezel AF, Ross JS, Stephens PJ, Ali SM,
JB, Spits H, van der Burg SH, Schumacher TN. High-throughput epitope Miller VA, Fakih M, Klempner SJ. Beyond microsatellite testing: assessment
discovery reveals frequent recognition of neo-antigens by CD4+ T cells in of tumor mutational burden identifies subsets of colorectal cancer who
human melanoma. Nat Med. 2015;21:81–5. may respond to immune checkpoint inhibition. J Gastrointest Oncol.
18. Snyder A, Makarov V, Merghoub T, Yuan J, Zaretsky JM, Desrichard A, 2018;9:610–7.
Walsh LA, Postow MA, Wong P, Ho TS, Hollmann TJ, Bruggeman C, Kannan 28. Vanderwalde A, Spetzler D, Xiao N, Gatalica Z, Marshall J. Microsatel-
K, Li Y, Elipenahli C, Liu C, Harbison CT, Wang L, Ribas A, Wolchok JD, Chan lite instability status determined by next-generation sequencing and
TA. Genetic basis for clinical response to CTLA-4 blockade in melanoma. compared with PD-L1 and tumor mutational burden in 11,348 patients.
N Engl J Med. 2014;371:2189–99. Cancer Med. 2018;7:746–56.
19. Devarakonda S, Rotolo F, Tsao MS, Lanc I, Brambilla E, Masood A, Olaussen 29. Le DT, Durham JN, Smith KN, Wang H, Bartlett BR, Aulakh LK, Lu S,
KA, Fulton R, Sakashita S, McLeer-Florin A, Ding K, Le Teuff G, Shepherd Kemberling H, Wilt C, Luber BS, Wong F, Azad NS, Rucki AA, Laheru D,
FA, Pignon JP, Graziano SL, Kratzke R, Soria JC, Seymour L, Govindan R, Donehower R, Zaheer A, Fisher GA, Crocenzi TS, Lee JJ, Greten TF, Duffy
Michiels S. Tumor mutation burden as a biomarker in resected non-small- AG, Ciombor KK, Eyring AD, Lam BH, Joe A, Kang SP, Holdhoff M, Danilova
cell lung cancer. J Clin Oncol. 2018;36:2995–3006. L, Cope L, Meyer C, Zhou S, Goldberg RM, Armstrong DK, Bever KM, Fader
20. Rizvi NA, Hellmann MD, Snyder A, Kvistborg P, Makarov V, Havel JJ, Lee AN, Taube J, Housseau F, Spetzler D, Xiao N, Pardoll DM, Papadopoulos N,
W, Yuan J, Wong P, Ho TS, Miller ML, Rekhtman N, Moreira AL, Ibrahim Kinzler KW, Eshleman JR, Vogelstein B, Anders RA, Diaz LA Jr. Mismatch
F, Bruggeman C, Gasmi B, Zappasodi R, Maeda Y, Sander C, Garon EB, repair deficiency predicts response of solid tumors to PD-1 blockade.
Merghoub T, Wolchok JD, Schumacher TN, Chan TA. Cancer immunology. Science. 2017;357:409–13.
Mutational landscape determines sensitivity to PD-1 blockade in non- 30. Rizvi H, Sanchez-Vega F, La K, Chatila W, Jonsson P, Halpenny D, Plod-
small cell lung cancer. Science. 2015;348:124–8. kowski A, Long N, Sauter JL, Rekhtman N, Hollmann T, Schalper KA, Gainor
JF, Shen R, Ni A, Arbour KC, Merghoub T, Wolchok J, Snyder A, Chaft JE,
Galuppini et al. Cancer Cell Int (2019) 19:209 Page 10 of 10

Kris MG, Rudin CM, Socci ND, Berger MF, Taylor BS, Zehir A, Solit DB, Arcila 36. Stadler ZK, Battaglin F, Middha S, Hechtman JF, Tran C, Cercek A, Yaeger
ME, Ladanyi M, Riely GJ, Schultz N, Hellmann MD. Molecular determinants R, Segal NH, Varghese AM, Reidy-Lagunes DL, Kemeny NE, Salo-Mullen
of response to anti-programmed cell death (PD)-1 and anti-programmed EE, Ashraf A, Weiser MR, Garcia-Aguilar J, Robson ME, Offit K, Arcila ME,
death-ligand 1 (PD-L1) blockade in patients with non-small-cell lung Berger MF, Shia J, Solit DB, Saltz LB. Reliable detection of mismatch repair
cancer profiled with targeted next-generation sequencing. J Clin Oncol. deficiency in colorectal cancers using mutational load in next-generation
2018;36:633–41. sequencing panels. J Clin Oncol. 2016;34:2141–7.
31. VanderLaan PA, Rangachari D, Costa DB. Lung cancer with a high tumor 37. Goodman AM, Kato S, Bazhenova L, Patel SP, Frampton GM, Miller V,
mutational burden. N Engl J Med. 2018;379:1093. Stephens PJ, Daniels GA, Kurzrock R. Tumor mutational burden as an
32. VanderLaan PA, Chen Y, DiStasio M, Rangachari D, Costa DB, Heher YK. independent predictor of response to immunotherapy in diverse cancers.
Molecular testing turnaround time in non-small-cell lung cancer: moni- Mol Cancer Ther. 2017;16:2598–608.
toring a moving target. Clin Lung Cancer. 2018;19:e589–90. 38. Fassan M. Molecular diagnostics in pathology: time for a next-generation
33. Lindeman NI, Cagle PT, Aisner DL, Arcila ME, Beasley MB, Bernicker EH, pathologist? Arch Pathol Lab Med. 2018;142:313–20.
Colasacco C, Dacic S, Hirsch FR, Kerr K, Kwiatkowski DJ, Ladanyi M, Nowak 39. VanderLaan PA, Rangachari D, Majid A, Parikh MS, Gangadharan SP, Kent
JA, Sholl L, Temple-Smolkin R, Solomon B, Souter LH, Thunnissen E, Tsao MS, McDonald DC, Huberman MS, Kobayashi SS, Costa DB. Tumor bio-
MS, Ventura CB, Wynes MW, Yatabe Y. Updated molecular testing guide- marker testing in non-small-cell lung cancer: a decade of change. Lung
line for the selection of lung cancer patients for treatment with targeted Cancer. 2018;116:90–5.
tyrosine kinase inhibitors: guideline from the college of american pathol- 40. Hellmann MD, Ciuleanu TE, Pluzanski A, Lee JS, Otterson GA, Audigier-
ogists, the international association for the study of lung cancer, and the Valette C, Minenza E, Linardou H, Burgers S, Salman P, Borghaei H, Rama-
association for molecular pathology. J Mol Diagn. 2018;20:129–59. lingam SS, Brahmer J, Reck M, O’Byrne KJ, Geese WJ, Green G, Chang H,
34. Meléndez B, Van Campenhout C, Rorive S, Remmelink M, Salmon I, Szustakowski J, Bhagavatheeswaran P, Healey D, Fu Y, Nathan F, Paz-Ares L.
D’Haene N. Methods of measurement for tumor mutational burden in Nivolumab plus ipilimumab in lung cancer with a high tumor mutational
tumor tissue. Transl Lung Cancer Res. 2018;7:661–7. burden. N Engl J Med. 2018;378:2093–104.
35. Chalmers ZR, Connelly CF, Fabrizio D, Gay L, Ali SM, Ennis R, Schrock A,
Campbell B, Shlien A, Chmielecki J, Huang F, He Y, Sun J, Tabori U, Ken-
nedy M, Lieber DS, Roels S, White J, Otto GA, Ross JS, Garraway L, Miller Publisher’s Note
VA, Stephens PJ, Frampton GM5. Analysis of 100,000 human cancer Springer Nature remains neutral with regard to jurisdictional claims in pub-
genomes reveals the landscape of tumor mutational burden. Genome lished maps and institutional affiliations.
Med. 2017;9:34.

Ready to submit your research ? Choose BMC and benefit from:

• fast, convenient online submission


• thorough peer review by experienced researchers in your field
• rapid publication on acceptance
• support for research data, including large and complex data types
• gold Open Access which fosters wider collaboration and increased citations
• maximum visibility for your research: over 100M website views per year

At BMC, research is always in progress.

Learn more biomedcentral.com/submissions

You might also like