Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Acta Oncologica

ISSN: 0284-186X (Print) 1651-226X (Online) Journal homepage: https://www.tandfonline.com/loi/ionc20

Prognostic role of lactate dehydrogenase in solid


tumors: A systematic review and meta-analysis of
76 studies

Fausto Petrelli, Mary Cabiddu, Andrea Coinu, Karen Borgonovo, Mara


Ghilardi, Veronica Lonati & Sandro Barni

To cite this article: Fausto Petrelli, Mary Cabiddu, Andrea Coinu, Karen Borgonovo, Mara Ghilardi,
Veronica Lonati & Sandro Barni (2015) Prognostic role of lactate dehydrogenase in solid tumors:
A systematic review and meta-analysis of 76 studies, Acta Oncologica, 54:7, 961-970, DOI:
10.3109/0284186X.2015.1043026

To link to this article: https://doi.org/10.3109/0284186X.2015.1043026

View supplementary material Published online: 18 May 2015.

Submit your article to this journal Article views: 5868

View related articles View Crossmark data

Citing articles: 57 View citing articles

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=ionc20
Acta Oncologica, 2015; 54: 961–970

REVIEW ARTICLE

Prognostic role of lactate dehydrogenase in solid tumors:


A systematic review and meta-analysis of 76 studies

FAUSTO PETRELLI, MARY CABIDDU, ANDREA COINU, KAREN BORGONOVO,


MARA GHILARDI, VERONICA LONATI & SANDRO BARNI

Department of Oncology, Medical Oncology Unit, Azienda Ospedaliera Treviglio, Treviglio (BG), Italy

ABSTRACT
Background. In cancer cells, metabolism is shifted to aerobic glycolysis with lactate production coupled with a higher
uptake of glucose as the main energy source. Lactate dehydrogenase (LDH) catalyzes the reduction of pyruvate to form
lactate, and serum level is often raised in aggressive cancer and hematological malignancies. We have assessed the
prognostic value of LDH in solid tumors.
Material and methods. A systematic review of electronic databases was conducted to identify publications exploring
the association of LDH with clinical outcome in solid tumors. Overall survival (OS) was the primary outcome, and
cancer-specific survival (CSS), progression-free survival (PFS), and disease-free survival (DFS) were secondary outcomes.
Data from studies reporting a hazard ratio (HR) and 95% confidence interval (CI) were pooled in a meta-analysis. Pooled
HRs were computed and weighted using generic inverse-variance and random-effect modeling. All statistical tests were
two-sided.
Results. Seventy-six studies comprising 22 882 patients, mainly with advanced disease, were included in the analysis.
Median cut-off of serum LDH was 245 U/L. Overall, higher LDH levels were associated with a HR for OS of 1.7 (95%
CI 1.62–1.79; p ⬍ 0.00001) in 73 studies. The prognostic effect was highest in renal cell, melanoma, gastric, prostate,
nasopharyngeal and lung cancers (all p ⬍ 0.00001). HRs for PFS was 1.75 (all p ⬍ 0.0001).
Conclusions. A high serum LDH level is associated with a poor survival in solid tumors, in particular melanoma, prostate
and renal cell carcinomas, and can be used as a useful and inexpensive prognostic biomarker in metastatic carcinomas.

The different metabolism of cancer compared with that has the highest efficiency to catalyze the conversion of
of normal cells confers a selective advantage for their pyruvate to lactate. LDH-5 is mainly localized to the
proliferation and survival. The use of lactate as an cytoplasm, where it participates in glucose metabolism.
energy source requires the conversion of lactate into Expression of LDHA and increased levels of function-
pyruvate as well as the transport of lactate into and out ally active LDH-5 are commonly observed events in
of tumor cells by specific transporters.The former path- highly invasive hypoxic cancers that are resistant to
way is regulated by lactate dehydrogenase (LDH). chemo- and radiotherapy. Biochemical markers of
LDHs catalyze the conversion of pyruvate and lactate, tumor burden, such as LDH have been incorporated
with concomitant conversion of NADH and NAD⫹. into several prognostic scores and staging for renal cell
These enzymes are homo- or hetero-tetramers com- carcinoma, melanoma and colorectal cancer [1–3]. In
posed of a M and a H protein subunits that are encoded metastatic germ cell tumors, furthermore, LDH is
by LDHA and LDHB genes, respectively. LDHA and incorporated in the IGCCCG prognostic classification,
pyruvate dehydrogenase kinase (PDK) are up-regulated and this possibly aids in guiding treatment and stratify-
in solid tumors in response to hypoxia in a HIF-1α- ing patients in clinical trials [4].
dependent manner. LDHA reducing the pyruvate into The significance and magnitude of the prognostic
lactate regenerates the NAD⫹ pool necessary to main- impact of circulating LDH are, however, unclear.
tain an adequate glycolytic process. The combinations LDH might be a simple and inexpensive objective
of LDHA and LDHB proteins into tetramers result in prognostic parameter that could be used in daily
the five major isoforms of LDH, numbered 1–5. LDH-5 oncologic clinical practice, other than helping to

Correspondence: F. Petrelli, Department of Oncology, Medical Oncology Unit, Azienda Ospedaliera Treviglio, Piazzale Ospedale 1, 24047, Treviglio (BG),
Italy. Tel: ⫹ 39 0363424420. Fax: ⫹ 39 0363424380. E-mail: faupe@libero.it

(Received 28 February 2015 ; accepted 14 April 2015)


ISSN 0284-186X print/ISSN 1651-226X online © 2015 Informa Healthcare
DOI: 10.3109/0284186X.2015.1043026
962 F. Petrelli et al.
stratify patients in clinical trials. The aim of this study Data extraction
was to summarize all data to quantify the prognostic
OS was the primary outcome of interest. CSS, PFS,
value of LDH on the clinical outcome in various solid
and DFS were secondary outcomes. The following
tumors, using standard meta-analysis techniques.
details were extracted: name of first author, type of
study, year of publication, number of patients
Material and methods included in the analysis, disease site, disease stage
This analysis was conducted in line with the Pre- (non-metastatic, metastatic, mixed [non-metastatic
ferred Reporting Items for Systematic Reviews and and metastatic]), cut-off defining high LDH, and
Meta-Analyses guidelines [5]. HRs for OS, PFS, DFS, or CSS as applicable. HRs
were extracted only from multivariable analyses.
Search methods and criteria for selecting studies for this When two or more HRs were reported for different
review cut-off values, only those above the median LDH
value for that study were considered.
An electronic search of PubMed, EMBASE,
SCOPUS, Web of Science, CINAHL and the Coch-
rane Register of Controlled Trials was performed. Data collection and statistical analysis
Search terms included: (“neoplasms”[MeSH Terms] The meta-analysis was conducted initially for all
OR “neoplasms”[All Fields] OR “cancer”[All Fields]) included studies for each of the endpoints of interest.
AND (LDH[All Fields] OR (“l-lactate dehydro- Subgroup analyses were conducted for disease site
genase”[MeSH Terms] OR (“l-lactate”[All Fields] and stage of disease only for the primary outcome.
AND “dehydrogenase”[All Fields]) OR “l-lactate Extracted data were aggregated into a meta-analysis
dehydrogenase”[All Fields] OR (“lactate”[All Fields] using RevMan 5.3 software (Cochrane Collabora-
AND “dehydrogenase”[All Fields]) OR “lactate tion, Copenhagen, Denmark). Estimates of HRs
dehydrogenase ” [All Fields])) AND ( “ mortality ” were weighted and pooled using the generic inverse-
[Subheading] OR “mortality”[All Fields] OR variance and random-effect model [6]. Analyses were
“survival”[All Fields] OR “survival”[MeSH Terms]) conducted for all studies, and differences between
AND (multivariate[All Fields] OR (cox[All Fields] diseases were assessed using methods described by
AND (“regression (psychology)”[MeSH Terms] OR Deeks et al. [7]. Meta-regression analysis was per-
(“regression”[All Fields] AND “(psychology)”[All formed to evaluate the effect of LDH cut-off level
Fields]) OR “regression (psychology)”[All Fields] (when available) on the HR for OS. Publication bias
OR “regression”[All Fields]))) AND (hazard[All for the primary endpoint was assessed by visual
Fields] AND (“Ratio (Oxf)”[Journal] OR “ratio”[All inspection of the funnel plot. Heterogeneity was
Fields])). Citation lists of retrieved articles were assessed with the use of the Cochran Q and I2 sta-
screened manually to ensure the sensitivity of the tistics. All statistical tests were two-sided, and statis-
search strategy. tical significance was defined as p ⬍ 0.05.
Inclusion criteria for the primary analysis were as
follows: 1) studies published in complete papers and
in the English language, of (at least 10) adult patients
Results
with solid tumors reporting on the prognostic impact
of serum LDH; and 2) availability of a hazard ratio Seventy-six studies [8–83] with a total of 22 882
(HR) and 95% confidence interval (CI) for overall patients were included (Figure 1). Characteristics of
survival (OS). For a secondary analysis, studies pro- the included studies are shown in (Supplementary
viding an HR for cancer-specific survival (CSS), Table I available online at http://informahealthcare.
progression-free survival (PFS) or time to progres- com/doi/abs/10.3109/0284186X.2015.1043026);
sion, and disease-free survival (DFS), were included most (95%) were published in the last decade. Among
as well. Duplicate publications were excluded. Two trials, eight studies regarded gastrointestinal malig-
reviewers (F. Petrelli, M. Cabiddu) evaluated inde- nancies (4 biliopancreatic, 2 gastric, 1 colorectal and
pendently all of the titles identified by the search 1 neuroendocrine cancers), seven head and neck can-
strategy. The results were then pooled, and all poten- cers, seven lung cancers, 12 melanomas, one sarcoma,
tially relevant publications were retrieved in full. The one thymic carcinoma, one mesothelioma, one breast
same two reviewers then evaluated the complete cancer, four mixed or unknown primaries, 16 prostate
articles for eligibility. To avoid inclusion of duplicated cancers, 15 renal cell carcinomas and three testicular
or overlapping data, we compared author names and cancers. Number of patients ranged from 35 to 2425.
institutions where patients were recruited and if sub- In all trials patients with mainly metastatic or locally
stantial doubts remained, the study reporting fewer advanced unresectable disease were included (only
patients was not included in the analysis. 5 trials included stage I–III cancers).
Prognostic role of LDH in solid tumors 963

(HR ⫽ 1.74; 95% CI 1.52–1.99, p ⬍ 0.00001), and


lung cancer (HR ⫽ 1.54; 95% CI 1.33–1.78,
p ⬍ 0.00001). The HR for the subgroup of mixed
solid tumors series or unknown primary was 1.74
(95% CI 1.4–2.15, p ⬍ 0.00001). Differences between
disease subgroups were statistically significant (p for
subgroup difference ⬍ 0.00001). Neuroendocrine
tumors, breast cancer, thymic carcinoma, mesothe-
lioma, testicular and colorectal cancer, and sarcoma
data were provided only in one paper each.
For the 15 disease-site subgroups analyzed, there
was statistically significant heterogeneity among trials
of biliopancreatic cancer, renal cell carcinoma, pros-
tate cancer, and melanoma, whereas heterogeneity
among other trials was non-statistically significant.
The scatter plot for the meta-regression is shown in
(Supplementary Figure 1 available online at http://
informahealthcare.com/doi/abs/10.3109/0284186X.
2015.1043026). Overall, there was a nearly statistically
significant association between LDH cut-off and the
HR for OS (p ⫽ 0.11). There was evidence of signifi-
cant publication bias, with several studies reporting
significantly higher results than expected (Figure 3).

Cancer-specific survival
Few studies reported this data, so a formal meta-
analysis was not performed.

Progression-free survival or time to progression


Thirteen trials reported HRs for PFS or TTP. Over-
all, a high LDH level was associated with a poor PFS
(HR ⫽ 1.75, 95% CI 1.31–2.33, p ⬍ 0.0001; I2 ⫽ 86%,
p for heterogeneity ⬍ 0.0001, random-effect model).
Figure 1. Study flow diagram of included studies.

Overall survival Disease-free survival


Seventy-three studies reported HR for OS. Forty- Only two studies including nasopharyngeal cancer
one studies did not specify a numerical cut-off used patients reported HRs for DFS. A high LDH con-
for multivariate analysis; conversely, in 34 studies a centration was associated with a lower DFS as mul-
predefined cut-off was reported (median: 245 U/L). tivariate analysis (HR ⫽ 1.68, 95% CI 1.24–2.27,
Twelve of the eligible 73 studies (16%) reported a p ⫽ 0.001; I2 ⫽ 47%, p for heterogeneity ⫽ 0.16, fixed
non-statistically significant HR. effect model).
Overall, LDH level was associated with a HR for
OS of 1.7 (95% CI 1.62–1.79; p ⬍ 0.00001; I2 ⫽ 92%,
Discussion
p for heterogeneity ⬍ 0.00001, random-effect model).
The effect of LDH level on OS among disease Several studies have suggested that elevated LDH is
subgroups is shown in Figure 2. The prognostic effect both a hallmark of aggressive carcinomas and lym-
of serum LDH, among subgroups with two or more phomas and associated with an unfavorable outcome.
studies, was highest in renal cell carcinoma (HR ⫽ 2.08; Indeed, LDH levels are actually included in the
95% CI 1.74–2.5, p ⬍ 0.00001), followed by mela- TNM staging system for melanoma [3], defining the
noma (HR ⫽ 1.97; 95% CI 1.59–2.43, p ⬍ 0.00001), M1c subgroup that is associated with a five-year OS
gastric cancer (HR ⫽ 1.91; 95% CI 1.38–2.63, of 10%. LDH is also considered with respect to the
p ⬍ 0.0001), prostate cancer (HR ⫽ 1.9; 95% CI risk status of testicular cancer, in particular meta-
1.58–2.29, p ⬍ 0.00001), nasopharyngeal carcinoma static non-seminoma germ cell tumors [84].
964 F. Petrelli et al.
Hazard Ratio Hazard Ratio
Study or Subgroup log[Hazard Ratio] SE Weight IV, Random, 95% CI Year IV, Random, 95% CI
1.1.1 Pancreatic-biliary
Masaki 2005 0.01 0.003 4.7% 1.01 [1.00, 1.02] 2005
Furuse 2009 0.615 0.304 0.6% 1.85 [1.02, 3.36] 2009
Nakai 2010 0.255 0.115 2.4% 1.29 [1.03, 1.62] 2010
Haas 2013 0.191 0.192 1.3% 1.21 [0.83, 1.76] 2013
Subtotal (95% CI) 9.0% 1.19 [0.96, 1.48]
Heterogeneity: Tau² = 0.03; Chi² = 9.38, df = 3 (P = 0.02); I² = 68%
Test for overall effect: Z = 1.59 (P = 0.11)

1.1.2 Gastric
Sougioultzis 2011 0.542 0.129 2.1% 1.72 [1.34, 2.21] 2011
Zhao 2014 0.907 0.273 0.7% 2.48 [1.45, 4.23] 2014
Subtotal (95% CI) 2.8% 1.91 [1.38, 2.63]
Heterogeneity: Tau² = 0.02; Chi² = 1.46, df = 1 (P = 0.23); I² = 32%
Test for overall effect: Z = 3.93 (P < 0.0001)

1.1.3 Colorectal
Chibaudel 2011 0.588 0.11 2.5% 1.80 [1.45, 2.23] 2011
Subtotal (95% CI) 2.5% 1.80 [1.45, 2.23]
Heterogeneity: Not applicable
Test for overall effect: Z = 5.35 (P < 0.00001)

1.1.4 Neuroendocrine
Yamaguchi 2014 0.43 0.19 1.3% 1.54 [1.06, 2.23] 2014
Subtotal (95% CI) 1.3% 1.54 [1.06, 2.23]
Heterogeneity: Not applicable
Test for overall effect: Z = 2.26 (P = 0.02)

1.1.5 Nasopharyngeal
Zhou 2011 1.111 0.317 0.6% 3.04 [1.63, 5.65] 2011
Li 2012 0.507 0.238 0.9% 1.66 [1.04, 2.65] 2012
Jin 2013 0.464 0.079 3.2% 1.59 [1.36, 1.86] 2013
Wan 2013 0.966 0.305 0.6% 2.63 [1.45, 4.78] 2013
Tian 2013 0.737 0.318 0.6% 2.09 [1.12, 3.90] 2013
Zeng 2014 0.522 0.179 1.4% 1.69 [1.19, 2.39] 2014
Wei 2014 0.545 0.176 1.4% 1.72 [1.22, 2.44] 2014
Subtotal (95% CI) 8.7% 1.74 [1.52, 1.99]
Heterogeneity: Tau² = 0.00; Chi² = 6.53, df = 6 (P = 0.37); I² = 8%
Test for overall effect: Z = 8.03 (P < 0.00001)

1.1.6 Lung
Ando 2004 0.449 0.483 0.3% 1.57 [0.61, 4.04] 2004
de Jong 2007 0.3 0.1 2.7% 1.35 [1.11, 1.64] 2007
Almasi 2012 0.693 0.261 0.8% 2.00 [1.20, 3.34] 2012
Lee 2014 1.319 0.634 0.2% 3.74 [1.08, 12.96] 2014
Kang 2014 0.41 0.171 1.5% 1.51 [1.08, 2.11] 2014
Wang 2014 0.594 0.114 2.4% 1.81 [1.45, 2.26] 2014
Ulas 2014 0.27 0.138 2.0% 1.31 [1.00, 1.72] 2014
Subtotal (95% CI) 9.8% 1.54 [1.33, 1.78]
Heterogeneity: Tau² = 0.01; Chi² = 8.06, df = 6 (P = 0.23); I² = 26%
Test for overall effect: Z = 5.70 (P < 0.00001)

1.1.7 Renal cell


Atzpodien 2003 0.262 0.134 2.0% 1.30 [1.00, 1.69] 2003
Peccatori 2005 0.888 0.335 0.5% 2.43 [1.26, 4.69] 2005
Donskov 2006 1.131 0.37 0.4% 3.10 [1.50, 6.40] 2006
Escudier 2007 0.519 0.189 1.3% 1.68 [1.16, 2.43] 2007
Richey 2010 0.438 0.194 1.3% 1.55 [1.06, 2.27] 2010
Vermaat 2010 1.281 0.383 0.4% 3.60 [1.70, 7.63] 2010
Culp 2010 0.507 0.141 1.9% 1.66 [1.26, 2.19] 2010
Abel 2011 0.884 0.333 0.5% 2.42 [1.26, 4.65] 2011
Patil 2011 0.658 0.109 2.5% 1.93 [1.56, 2.39] 2011
Armstrong 2012 1.033 0.171 1.5% 2.81 [2.01, 3.93] 2012
Kamba 2014 1.176 0.551 0.2% 3.24 [1.10, 9.54] 2014
Cetin 2014 0.801 0.374 0.4% 2.23 [1.07, 4.64] 2014
Atkinson 2014 1.115 0.342 0.5% 3.05 [1.56, 5.96] 2014
Amato 2014 1.91 0.663 0.1% 6.75 [1.84, 24.77] 2014
Subtotal (95% CI) 13.6% 2.08 [1.74, 2.50]
Heterogeneity: Tau² = 0.05; Chi² = 28.03, df = 13 (P = 0.009); I² = 54%
Test for overall effect: Z = 7.90 (P < 0.00001)

1.1.8 Testicular
Gerlinger 2010 1.224 0.354 0.5% 3.40 [1.70, 6.81] 2010
Subtotal (95% CI) 0.5% 3.40 [1.70, 6.81]
Heterogeneity: Not applicable
Test for overall effect: Z = 3.46 (P = 0.0005)

1.1.9 Prostate
D'amico 2005 1.03 0.173 1.5% 2.80 [2.00, 3.93] 2005
George 2005 0.3 0.156 1.7% 1.35 [0.99, 1.83] 2005
Taplin 2005 0.473 0.118 2.3% 1.60 [1.27, 2.02] 2005
Humphrey 2006 0.306 0.172 1.5% 1.36 [0.97, 1.90] 2006
Saito 2007 0.482 0.236 0.9% 1.62 [1.02, 2.57] 2007
Scher 2009 1.335 0.225 1.0% 3.80 [2.44, 5.91] 2009
Coumans 2010 0.187 0.24 0.9% 1.21 [0.75, 1.93] 2010
Yamada 2011 0.902 0.39 0.4% 2.46 [1.15, 5.29] 2011
Kume 2011 0.358 1.087 0.1% 1.43 [0.17, 12.04] 2011
Narita 2012 1.806 0.666 0.1% 6.09 [1.65, 22.45] 2012
Armstrong 2013 0.56 0.246 0.9% 1.75 [1.08, 2.84] 2013
Omlin 2013 0.555 0.139 1.9% 1.74 [1.33, 2.29] 2013
Sonpavde 2014 1.051 0.113 2.4% 2.86 [2.29, 3.57] 2014
Halabi 2014 0.336 0.096 2.8% 1.40 [1.16, 1.69] 2014
Templeton 2014 0.924 0.213 1.1% 2.52 [1.66, 3.82] 2014
Ferraldeschi 2014 0.464 0.237 0.9% 1.59 [1.00, 2.53] 2014
Subtotal (95% CI) 20.4% 1.90 [1.58, 2.29]
Heterogeneity: Tau² = 0.09; Chi² = 58.03, df = 15 (P < 0.00001); I² = 74%
Test for overall effect: Z = 6.82 (P < 0.00001)

1.1.10 Melanoma
Franzke 1998 0.494 0.132 2.1% 1.64 [1.27, 2.12] 1998
Chiarion-Sileni 2003 1.163 0.222 1.0% 3.20 [2.07, 4.94] 2003
Schmidt 2005 1.03 0.123 2.2% 2.80 [2.20, 3.56] 2005
Soubrane 2005 0.52 0.174 1.5% 1.68 [1.20, 2.37] 2005
Schmidt 2007 0.788 0.195 1.2% 2.20 [1.50, 3.22] 2007
Neuman 2008 0.351 0.126 2.2% 1.42 [1.11, 1.82] 2008
Staudt 2010 1.988 0.347 0.5% 7.30 [3.70, 14.41] 2010
Jakob 2012 1.012 0.2 1.2% 2.75 [1.86, 4.07] 2012
Weide 2012 0.47 0.106 2.6% 1.60 [1.30, 1.97] 2012
Wevers 2013 0.329 0.315 0.6% 1.39 [0.75, 2.58] 2013
Gray 2014 0.399 0.102 2.7% 1.49 [1.22, 1.82] 2014
Gaudy-Marqueste 2014 -0.144 0.341 0.5% 0.87 [0.44, 1.69] 2014
Subtotal (95% CI) 18.2% 1.97 [1.59, 2.43]
Heterogeneity: Tau² = 0.10; Chi² = 55.47, df = 11 (P < 0.00001); I² = 80%
Test for overall effect: Z = 6.28 (P < 0.00001)

1.1.11 Sarcoma
Gonzalez-Billalabeitia 2009 2.239 0.862 0.1% 9.38 [1.73, 50.83] 2009
Subtotal (95% CI) 0.1% 9.38 [1.73, 50.83]
Heterogeneity: Not applicable
Test for overall effect: Z = 2.60 (P = 0.009)

1.1.12 Mixed or unknown


Lagerwaard 1999 0.438 0.082 3.2% 1.55 [1.32, 1.82] 1999
Trivanovic 2009 0.793 0.229 1.0% 2.21 [1.41, 3.46] 2009
Tredan 2011 0.47 0.164 1.6% 1.60 [1.16, 2.21] 2011
Pinato 2014 1.163 0.422 0.3% 3.20 [1.40, 7.32] 2014
Subtotal (95% CI) 6.0% 1.74 [1.40, 2.15]
Heterogeneity: Tau² = 0.02; Chi² = 4.71, df = 3 (P = 0.19); I² = 36%
Test for overall effect: Z = 5.03 (P < 0.00001)

1.1.13 Mesothelioma
Najmi 2014 0.01 0.005 4.7% 1.01 [1.00, 1.02] 2014
Subtotal (95% CI) 4.7% 1.01 [1.00, 1.02]
Heterogeneity: Not applicable
Test for overall effect: Z = 2.00 (P = 0.05)

1.1.14 Thymic carcinoma


Wu 2014 1.025 0.361 0.4% 2.79 [1.37, 5.66] 2014
Subtotal (95% CI) 0.4% 2.79 [1.37, 5.66]
Heterogeneity: Not applicable
Test for overall effect: Z = 2.84 (P = 0.005)

1.1.15 Breast
Liu 2014 0.351 0.14 1.9% 1.42 [1.08, 1.87] 2014
Subtotal (95% CI) 1.9% 1.42 [1.08, 1.87]
Heterogeneity: Not applicable
Test for overall effect: Z = 2.51 (P = 0.01)

Total (95% CI) 100.0% 1.70 [1.62, 1.79]


Heterogeneity: Tau² = 0.01; Chi² = 922.95, df = 72 (P < 0.00001); I² = 92%
Test for overall effect: Z = 20.98 (P < 0.00001) 0.02 0.1 1 10 50
Test for subgroup differences: Chi² = 335.54, df = 14 (P < 0.00001), I² = 95.8% Favours low LDH Favours high LDH

Figure 2. Forest plots showing hazard ratio for overall survival for LDH greater than or less than the cut-off for overall studies and by
disease subgroups.

We performed a meta-analysis of 76 studies found a consistent impact of elevated LDH on sur-


involving 22 882 patients with solid tumors in order vival (HR ⫽ 1.7) among various disease subgroups,
to assess the prognostic effect of serum LDH. We and mainly in advanced disease stages. High serum
Prognostic role of LDH in solid tumors 965
SE(log[Hazard Ratio])
0

0.5

1.5

Hazard Ratio
2
0.02 0.1 1 10 50
Subgroups
Pancreatic-biliary Nasopharyngeal Prostate Mesothelioma
Gastric Lung Melanoma Thymic carcinoma
Colorectal Renal cell Sarcoma Breast
Neuroendocrine Testicular Mixed or unknown

Figure 3. Funnel plot of hazard ratio for overall survival for serum LDH level (horizontal axis) and the standard error (SE) for the hazard
ratio (vertical axis). Each study is represented by one circle. The vertical line represents the pooled effect estimate.

LDH was also associated with a reduced PFS in 13 the other metastatic malignancies examined in this
trials. As expected, the magnitude of the effect on OS meta-analysis. It is possible that C-reactive protein
was the greatest in melanoma and renal cell carcinoma reflects the enhanced immune response elicited by a
other than metastatic (castration-resistant) prostate tumor, which is more intense when tissue necrosis
cancer. The burden of the data considered (about (and so LDH) and inflammation are high, as occurs
50% of the included studies) concerns these neoplas- with more aggressive cancer subtypes.
tic diseases, in which hypoxia and angiogenic factors That lactates play a role in cancer cells is unsurpris-
play a crucial role. In particular, in renal cell carci- ing, and LDH concentrations in human serum during
noma, a five-parameter-based prognostic score pro- neoplastic conditions can be regarded as a marker of
vided by the Memorial Sloan-Kettering Cancer (cancer) the metabolic activity and avid glucose uptake
Center that is commonly used in patients with meta- of cells. Tumor hypoxia actually induces, among other
static disease includes LDH levels and correlates these things, the expression of a hypoxia-inducible factor
with outcomes in patients treated in various clinical (HIF), which is a transcription factor that initiates a
trials before and after the era of targeted therapy range of activities, including angiogenesis and various
[85,86]. The present analysis mainly covered patients prosurvival mechanisms [95]. In terms of tumor growth,
with advanced tumors, which confirms that the prog- the local blood supply becomes inadequate, thus requir-
nostic role of LDH currently seems to be confined to ing angiogenesis and leading to hypoxia. The metabo-
only highly proliferative and metastatic cancers. We lism of cells is consecutively shifted towards the
focused on solid tumors, because the prognostic role anerobic glycolytic pathway (e.g. from glucose to
of LDH in hematological disease is well known, espe- lactate) through the enhanced expression of glycolytic
cially in high-grade lymphoma where it is elevated in enzymes and glucose transporters. This is coupled with
almost 50% of patients [87]. The novelty of our data a reduced dependence on the oxidative pathway. Can-
is that it particularly relates to prostate cancer, where cers are also able to produce lactate due to increased
the role of LDH as a prognostic factor has not yet glycolytic activity, regardless of oxygen availability (the
been fully demonstrated. Several papers reporting the so-called Warburg effect), which is a phenomenon that
role of inflammation parameters (neutrophil-lympo- was first described about a century ago as being a con-
cyte ratio and C-reactive protein) other than bone- sequence of energy requests from tumor cells. Lactate
related biomarkers (e.g. alkaline phosphatase) have which is the end product of glycolysis from pyruvate, is
recently been published [88–94]. In particular, LDH produced in large amounts in highly aggressive tumors
seems to be associated with a poor prognosis in met- in response to the specific characteristics of the microen-
astatic, castration-resistant prostate cancer, and could vironment and the genetic features of neoplastic cells.
thus be valuable for identifying patients who are suit- Importantly, lactate also constitutes an alternative met-
able for the earlier use of chemotherapy. Data on abolic source for cancer cells [96–98]. The LDH-A
LDH levels and inflammatory parameters could be gene promoter possesses two conserved hypoxia
useful when starting therapy for prostate cancer and response elements containing functionally-essential
966 F. Petrelli et al.
binding sites for HIF-1 with the consensus sequence Recent in vitro studies have shown that inhibiting
5´-RCGTG-3´, which may strongly suggest an oxygen- LDHA expression may lessen invasiveness and the
dependent regulation of LDH-5 activity [99]. metastatic potential of cancer cells by reducing their
Recently, high LDH levels have been found to proliferation capacity and reversing their resistance
predict the response to anti-angiogenetic agents, to chemotherapy [102–110]. LDH can thus be
such as bevacizumab and sorafenib. In two papers, regarded as an ideal target for anticancer therapies.
Scartozzi et al. examined this predictive role, dem- In conclusion, serum LDH is associated with poor
onstrating that: 1) in subjects with high serum LDH, survival with respect to many solid tumors, and could
bevacizumab confers a higher RR in colorectal be used as a cost-effective prognostic biomarker in
cancer; and 2) in hepatocellular carcinoma treated terms of prognostic scores before treating advanced
with sorafenib, LDH levels seem to predict clinical disease and enrolling patients in prospective trials.
outcomes in terms of PFS and OS [100,101]. The evaluation of LDH as a therapeutic target is also
This meta-analysis does, however, have some warranted, and is the subject of ongoing trials.
limitations. First, only published data, as opposed to
individual patient data, have been collected. Second,
Declaration of interest: The authors report no
we found evidence of high heterogeneity among the
conflicts of interest. The authors alone are respon-
trials, as well as a significant publication bias, with
sible for the content and writing of the paper.
a larger number of trials than expected reporting a
significantly higher effect of LDH levels. Further-
more, we only included studies reporting HRs, while References
more than 20 publications reporting on the prognos- [1] Motzer RJ, Mazumdar M, Bacik J, Berg W, Amsterdam A,
tic value of LDH were excluded (e.g. because only Ferrara J. Survival and prognostic stratification of 670
odds ratios, relative risk for survival, or p for sig- patients with advanced renal cell carcinoma. J Clin Oncol
1999;17:2530–40.
nificance were reported), possibly introducing bias. [2] Chibaudel B, Bonnetain F, Tournigand C, Bengrine-Lefevre L,
It is notable that almost all of the excluded papers Teixeira L, Artru P, et al. Simplified prognostic model in
(the majority analyzing genitourinary tumors, sarco- patients with oxaliplatin-based or irinotecan-based first-line
mas, melanomas and lung cancers) presented a sig- chemotherapy for metastatic colorectal cancer: A GERCOR
nificant association with outcome, thus reducing the study. Oncologist 2011;16:1228–38.
[3] Balch CM, Soong SJ, Atkins MB, Buzaid AC, Cascinelli N,
risk that could have changed the final results. Many Coit DG, et al. An evidence-based staging system for cuta-
other case series, however, were excluded because neous melanoma. CA Cancer J Clin 2004;54:131–49; quiz
serum LDH data was unavailable in the population 182–4.
analyzed. Third, LDH is influenced by other non- [4] International Germ Cell Consensus Classification: A prog-
neoplastic conditions, including heart failure, hypo- nostic factor-based staging system for metastatic germ cell
cancers. International Germ Cell Cancer Collaborative
thyroidism, anemia, and lung or liver disease, which Group. J Clin Oncol 1997;15:594–603.
may have influenced the serum concentrations. Most [5] Liberati A, Altman DG, Tetzlaff J, Mulrow C, Gøtzsche PC,
of the studies included in this meta-analysis did not Ioannidis JP, et al. The PRISMA statement for reporting
explicitly control for such different, benign condi- systematic reviews and meta-analyses of studies that evalu-
tions that may have affected LDH levels. Finally, a ate health care interventions: Explanation and elaboration.
PLoS Med 2009;6:e1000100.
cut-off value associated with a significantly worse [6] Cochrane handbook for systematic reviews of interventions.
OS cannot be defined, even if the median of the [cited 2014 Dec]. http://www.cochrane.org/training/cochrane-
reported cut-off values is the common upper normal handbook.
limit used in most laboratories (245 U/L). Unfortu- [7] Deeks JJ, Higgins JPT, Altman DG (editors). Chapter 9:
nately, only about 50% of the papers considered Analysing data and undertaking meta-analyses. In: Higgins
JPT, Green S (editors). Cochrane handbook for systematic
provided details of such a cut-off point, and the reviews of interventions version 5.1.0 [updated 2011 Mar;
metaregression for correlations of serum cut-off cited 2014 Dec]. The Cochrane Collaboration, 2011. Avail-
levels with HRs was not significant. However, to our able from: www.cochrane-handbook.org.
knowledge, this is the first meta-analysis to system- [8] Zhao Z, Han F, Yang S, Hua L, Wu J, Zhan W. The clinico-
atically confirm the poor prognostic value of high pathologic importance of serum lactic dehydrogenase in
patients with gastric cancer. Dis Markers 2014;140913.
LDH concentrations in solid tumors. It also involves [9] Wu JX, Chen HQ, Shao LD, Qiu SF, Ni QY, Zheng BH,
22 961 patients included in 76 published studies, et al. Long-term follow-up and prognostic factors for advanced
mainly concerning genitourinary and melanoma thymic carcinoma. Medicine (Baltimore) 2014;93:e324.
tumors. The value of the analysis does, however, [10] Liu X, Meng QH, Ye Y, Hildebrandt M, Gu J, Wu X. Prog-
remain confined to advanced tumors with locally nostic significance of pretreatment serum levels of albumin,
LDH and total bilirubin in patients with non-metastatic
advanced and metastatic disease, and its significance breast cancer. Carcinogenesis Epub 2015;36:243–8.
with respect to the early stages of disease requires [11] Najmi K, Khosravi A, Seifi S, Emami H, Chaibakhsh S,
further evaluation. Radmand G, et al. Clinicopathologic and survival charac-
Prognostic role of LDH in solid tumors 967
teristics of malignant pleural mesothelioma registered in dehydrogenase variability and mortality among metastatic
hospital cancer registry. Tanaffos 2014;13:6–12. renal cell carcinoma. Int Urol Nephrol 2014;46:1081–7.
[12] Ulas A, Turkoz FP, Silay K, Tokluoglu S, Avci N, [27] Armstrong AJ, Häggman M, Stadler WM, Gingrich JR,
Oksuzoglu B, et al. A laboratory prognostic index model for Assikis V, Polikoff J, et al. Long-term survival and biomarker
patients with advanced non-small cell lung cancer. PLoS correlates of tasquinimod efficacy in a multicenter rand-
One 2014;9:e114471. omized study of men with minimally symptomatic meta-
[13] Ferraldeschi R, Nava Rodrigues D, Riisnaes R, Miranda S, static castration-resistant prostate cancer. Clin Cancer Res
Figueiredo I, Rescigno P, et al. PTEN protein loss and clin- 2013;19:6891–901.
ical outcome from castration-resistant prostate cancer treated [28] Tian YM, Zeng L, Wang FH, Liu S, Guan Y, Lu TX, et al.
with abiraterone acetate. Eur Urol 2015;67:795–802. Prognostic factors in nasopharyngeal carcinoma with syn-
[14] Zeng L, Tian YM, Huang Y, Sun XM, Wang FH, Deng XW, chronous liver metastasis: A retrospective study for the
et al. Retrospective analysis of 234 nasopharyngeal carci- management of treatment. Radiat Oncol 2013;8:272.
noma patients with distant metastasis at initial diagnosis: [29] Gray MR, Martin del Campo S, Zhang X, Zhang H,
Therapeutic approaches and prognostic factors. PLoS One Souza FF, Carson WE 3rd, et al. Metastatic melanoma:
2014;9:e108070. Lactate dehydrogenase levels and CT imaging findings of
[15] Templeton AJ, Pezaro C, Omlin A, McNamara MG, tumor devascularization allow accurate prediction of sur-
Leibowitz-Amit R, Vera-Badillo FE, et al. Simple prognostic vival in patients treated with bevacizumab. Radiology
score for metastatic castration-resistant prostate cancer with 2014;270:425–34.
incorporation of neutrophil-to-lymphocyte ratio. Cancer [30] Atkinson BJ, Kalra S, Wang X, Bathala T, Corn P, Tannir NM,
2014;120:3346–52. et al. Clinical outcomes for patients with metastatic renal cell
[16] Yamaguchi T, Machida N, Morizane C, Kasuga A, carcinoma treated with alternative sunitinib schedules. J Urol
Takahashi H, Sudo K, et al. Multicenter retrospective analysis 2014;191:611–8.
of systemic chemotherapy for advanced neuroendocrine carci- [31] Kamba T, Yamasaki T, Teramukai S, Shibasaki N, Arakaki R,
noma of the digestive system. Cancer Sci 2014;105:1176–81. Sakamoto H, et al. Improvement of prognosis in patients with
[17] Kang MH, Go SI, Song HN, Lee A, Kim SH, Kang JH, metastatic renal cell carcinoma and Memorial Sloan-Kettering
et al. The prognostic impact of the neutrophil-to-lymphocyte Cancer Center intermediate risk features by modern strategy
ratio in patients with small-cell lung cancer. Br J Cancer including molecular-targeted therapy in clinical practice. Int J
2014;111:452–60. Clin Oncol 2014;19:505–15.
[18] Wei Z, Zeng X, Xu J, Duan X, Xie Y. Prognostic value of [32] Powles T, Bascoul-Mollevi C, Kramar A, Lorch A, Beyer J. Prog-
pretreatment serum levels of lactate dehydrogenase in non- nostic impact of LDH levels in patients with relapsed/refractory
metastatic nasopharyngeal carcinoma: Single-site analysis seminoma. J Cancer Res Clin Oncol 2013;139:1311–6.
of 601 patients in a highly endemic area. Oncol Targets Ther [33] Wan XB, Wei L, Li H, Dong M, Lin Q, Ma XK, et al. High
2014;7:739–49. pretreatment serum lactate dehydrogenase level correlates
[19] Sonpavde G, Pond GR, Armstrong AJ, Clarke SJ, Vardy JL, with disease relapse and predicts an inferior outcome in
Templeton AJ, et al. Prognostic impact of the neutrophil- locally advanced nasopharyngeal carcinoma. Eur J Cancer
to-lymphocyte ratio in men with metastatic castration-resistant 2013;49:2356–64.
prostate cancer. Clin Genitourin Cancer 2014;12:317–24. [34] Wevers KP, Kruijff S, Speijers MJ, Bastiaannet E,
[20] Wang X, Jiang R, Li K. Prognostic significance of pretreat- Muller Kobold AC, Hoekstra HJ. S-100B: A stronger prog-
ment laboratory parameters in combined small-cell lung nostic biomarker than LDH in stage IIIB-C melanoma.
cancer. Cell Biochem Biophys 2014;69:633–40. Ann Surg Oncol 2013;20:2772–9.
[21] Halabi S, Lin CY, Kelly WK, Fizazi KS, Moul JW, Kaplan EB, [35] Haas M, Heinemann V, Kullmann F, Laubender RP,
et al. Updated prognostic model for predicting overall survival Klose C, Bruns CJ, et al. Prognostic value of CA 19-9,
in first-line chemotherapy for patients with metastatic castra- CEA, CRP, LDH and bilirubin levels in locally advanced
tion-resistant prostate cancer. J Clin Oncol 2014;32:671–7. and metastatic pancreatic cancer: Results from a multi-
Erratum in: J Clin Oncol 2014;32:1387. center, pooled analysis of patients receiving palliative chem-
[22] Gaudy-Marqueste C, Archier E, Grob A, Durieux O, otherapy. J Cancer Res Clin Oncol 2013;139:681–9.
Loundou A, Richard MA, et al. Initial metastatic kinetics is [36] Omlin A, Pezaro C, Mukherji D, Mulick Cassidy A,
the best prognostic indicator in stage IV metastatic Sandhu S, Bianchini D, et al. Improved survival in a cohort
melanoma. Eur J Cancer 2014;50:1120–4. of trial participants with metastatic castration-resistant
[23] Pinato DJ, Stavraka C, Flynn MJ, Forster MD, O’Cathail SM, prostate cancer demonstrates the need for updated prog-
Seckl MJ, et al. An inflammation based score can optimize nostic nomograms. Eur Urol 2013;64:300–6.
the selection of patients with advanced cancer considered for [37] Jin Y,Ye X, Shao L, Lin BC, He CX, Zhang BB, et al. Serum
early phase clinical trials. PLoS One 2014;9:e83279. lactic dehydrogenase strongly predicts survival in metastatic
[24] Lee J, Kim JO, Jung CK, Kim YS, Yoo IeR, Choi WH, et al. nasopharyngeal carcinoma treated with palliative chemo-
Metabolic activity on [18f]-fluorodeoxyglucose-positron therapy. Eur J Cancer 2013;49:1619–26.
emission tomography/computed tomography and glucose [38] Almasi CE, Drivsholm L, Pappot H, Høyer-Hansen G,
transporter-1 expression might predict clinical outcomes in Christensen IJ. The liberated domain I of urokinase plas-
patients with limited disease small-cell lung cancer who minogen activator receptor – a new tumour marker in small
receive concurrent chemoradiation. Clin Lung Cancer cell lung cancer. APMIS 2013;121:189–96.
2014;15:e13–21. [39] Armstrong AJ, George DJ, Halabi S. Serum lactate dehydro-
[25] Amato RJ, Flaherty A, Zhang Y, Ouyang F, Mohlere V. genase predicts for overall survival benefit in patients with
Clinical prognostic factors associated with outcome in metastatic renal cell carcinoma treated with inhibition of mam-
patients with renal cell cancer with prior tyrosine kinase malian target of rapamycin. J Clin Oncol 2012;30:3402–7.
inhibitors or immunotherapy treated with everolimus. Urol [40] Weide B, Elsässer M, Büttner P, Pflugfelder A, Leiter U,
Oncol 2014;32:345–54. Eigentler TK, et al. Serum markers lactate dehydrogenase and
[26] Cetin B, Afsar B, Deger SM, Gonul II, Gumusay O, Ozet A, S100B predict independently disease outcome in melanoma
et al. Association between hemoglobin, calcium, and lactate patients with distant metastasis. Br J Cancer 2012;107:422–8.
968 F. Petrelli et al.
[41] Li G, Gao J, Tao YL, Xu BQ, Tu ZW, Liu ZG, et al. Increased static renal cell carcinoma for cytoreductive nephrectomy?
pretreatment levels of serum LDH and ALP as poor prog- Cancer 2010;116:3378–88.
nostic factors for nasopharyngeal carcinoma. Chin J Cancer [56] Nakai Y, Isayama H, Sasaki T, Sasahira N, Kogure H,
2012;31:197–206. Hirano K, et al. Impact of S-1 in patients with gemcitabine-
[42] Jakob JA, Bassett RL Jr, Ng CS, Curry JL, Joseph RW, refractory pancreatic cancer in Japan. Jpn J Clin Oncol
Alvarado GC, et al. NRAS mutation status is an independ- 2010;40:774–80.
ent prognostic factor in metastatic melanoma. Cancer [57] Staudt M, Lasithiotakis K, Leiter U, Meier F, Eigentler T,
2012;118:4014–23. Bamberg M, et al. Determinants of survival in patients with
[43] Zhou GQ, Tang LL, Mao YP, Chen L, Li WF, Sun Y, et al. brain metastases from cutaneous melanoma. Br J Cancer
Baseline serum lactate dehydrogenase levels for patients 2010;102:1213–8.
treated with intensity-modulated radiotherapy for nasopha- [58] Coumans FA, Doggen CJ, Attard G, de Bono JS,
ryngeal carcinoma: A predictor of poor prognosis and sub- Terstappen LW. All circulating EpCAM⫹ CK⫹ CD45- objects
sequent liver metastasis. Int J Radiat Oncol Biol Phys predict overall survival in castration-resistant prostate cancer.
2012;82:e359–65. Erratum in: Int J Radiat Oncol Biol Phys Ann Oncol 2010;21:1851–7.
2013;86:213. [59] Vermaat JS, van der Tweel I, Mehra N, Sleijfer S, Haanen JB,
[44] Chibaudel B, Bonnetain F, Tournigand C, Bengrine-Lefevre L, Roodhart JM, et al. Two-protein signature of novel serological
Teixeira L, Artru P, et al. Simplified prognostic model in markers apolipoprotein-A2 and serum amyloid alpha predicts
patients with oxaliplatin-based or irinotecan-based first-line prognosis in patients with metastatic renal cell cancer and
chemotherapy for metastatic colorectal cancer: A GERCOR improves the currently used prognostic survival models. Ann
study. Oncologist 2011;16:1228–38. Oncol 2010;21:1472–81.
[45] Abel EJ, Culp SH, Tannir NM, Tamboli P, Matin SF, [60] González-Billalabeitia E, Hitt R, Fernández J, Conde E,
Wood CG. Early primary tumor size reduction is an inde- Martínez-Tello F, Enríquez de Salamanca R, et al. Pre-
pendent predictor of improved overall survival in metastatic treatment serum lactate dehydrogenase level is an impor-
renal cell carcinoma patients treated with sunitinib. Eur tant prognostic factor in high-grade extremity osteosarcoma.
Urol 2011;60:1273–9. Clin Transl Oncol 2009;11:479–83.
[46] Narita S, Tsuchiya N, Yuasa T, Maita S, Obara T, [61] Furuse J, Okusaka T, Ohkawa S, Nagase M, Funakoshi A,
Numakura K, et al. Outcome, clinical prognostic factors Boku N, et al. A phase II study of uracil-tegafur plus doxo-
and genetic predictors of adverse reactions of intermittent rubicin and prognostic factors in patients with unresectable
combination chemotherapy with docetaxel, estramustine biliary tract cancer. Cancer Chemother Pharmacol
phosphate and carboplatin for castration-resistant prostate 2009;65:113–20.
cancer. Int J Clin Oncol 2012;17:204–11. [62] Scher HI, Jia X, de Bono JS, Fleisher M, Pienta KJ,
[47] Zhang HL, Zhu Y, Wang CF, Yao XD, Zhang SL, Dai B, Raghavan D, et al. Circulating tumour cells as prognostic
et al. Erythrocyte sedimentation rate kinetics as a marker markers in progressive, castration-resistant prostate cancer:
of treatment response and predictor of prognosis in Chinese A reanalysis of IMMC38 trial data. Lancet Oncol
metastatic renal cell carcinoma patients treated with soraf- 2009;10:233–9.
enib. Int J Urol 2011;18:422–30. [63] Trivanovi D, Petkovic M, Stimac D. New prognostic index
[48] Trédan O, Ray-Coquard I, Chvetzoff G, Rebattu P, to predict survival in patients with cancer of unknown pri-
Bajard A, Chabaud S, et al. Validation of prognostic scores mary site with unfavourable prognosis. Clin Oncol (R Coll
for survival in cancer patients beyond first-line therapy. Radiol) 2009;21:43–8.
BMC Cancer 2011;11:95. [64] Neuman HB, Patel A, Ishill N, Hanlon C, Brady MS,
[49] Kume H, Suzuki M, Fujimura T, Fukuhara H, Enomoto Y, Halpern AC, et al. A single-institution validation of the
Nishimatsu H, et al. Docetaxel as a vital option for corti- AJCC staging system for stage IV melanoma. Ann Surg
costeroid-refractory prostate cancer. Int Urol Nephrol Oncol 2008;15:2034–41.
2011;43:1081–7. [65] Saito T, Hara N, Kitamura Y, Komatsubara S. Prostate-
[50] Sougioultzis S, Syrios J, Xynos ID, Bovaretos N, Kosmas C, specific antigen/prostatic acid phosphatase ratio is signifi-
Sarantonis J, et al. Palliative gastrectomy and other factors cant prognostic factor in patients with stage IV prostate
affecting overall survival in stage IV gastric adenocarcinoma cancer. Urology 2007;70:702–5.
patients receiving chemotherapy: A retrospective analysis. [66] Escudier B, Choueiri TK, Oudard S, Szczylik C, Négrier S,
Eur J Surg Oncol 2011;37:312–8. Ravaud A, et al. Prognostic factors of metastatic renal cell
[51] Yamada Y, Nakamura K, Aoki S, Tobiume M, Zennami K, carcinoma after failure of immunotherapy: New paradigm
Kato Y, et al. Lactate dehydrogenase, Gleason score and from a large phase III trial with shark cartilage extract AE
HER-2 overexpression are significant prognostic factors for 941. J Urol 2007;178:1901–5.
M1b prostate cancer. Oncol Rep 2011;25:937–44. [67] de Jong WK, Fidler V, Groen HJ. Prognostic classification
[52] Richey SL, Culp SH, Jonasch E, Corn PG, Pagliaro LC, with laboratory parameters or imaging techniques in small-
Tamboli P, et al. Outcome of patients with metastatic renal cell lung cancer. Clin Lung Cancer 2007;8:376–81.
cell carcinoma treated with targeted therapy without cytore- [68] Humphrey PA, Halabi S, Picus J, Sanford B, Vogelzang NJ,
ductive nephrectomy. Ann Oncol 2011;22:1048–53. Small EJ, et al. Prognostic significance of plasma scatter factor/
[53] Gerlinger M, Wilson P, Powles T, Shamash J. Elevated LDH hepatocyte growth factor levels in patients with metastatic
predicts poor outcome of recurrent germ cell tumours treated hormone- refractory prostate cancer: Results from cancer and
with dose dense chemotherapy. Eur J Cancer 2010;46:2913–8. leukemia group B 150005/9480. Clin Genitourin Cancer
[54] Patil S, Figlin RA, Hutson TE, Michaelson MD, Négrier S, 2006;4:269–74.
Kim ST, et al. Prognostic factors for progression-free and [69] Donskov F, von der Maase H. Impact of immune parameters
overall survival with sunitinib targeted therapy and with on long-term survival in metastatic renal cell carcinoma. J
cytokine as first-line therapy in patients with metastatic Clin Oncol 2006;24:1997–2005.
renal cell carcinoma. Ann Oncol 2011; 22:295–300. [70] Schmidt H, Johansen JS, Gehl J, Geertsen PF, Fode K,
[55] Culp SH, Tannir NM, Abel EJ, Margulis V, Tamboli P, von der Maase H. Elevated serum level of YKL-40 is an
Matin SF, et al. Can we better select patients with meta- independent prognostic factor for poor survival in patients
Prognostic role of LDH in solid tumors 969
with metastatic melanoma. Cancer 2006;106:1130–9. [85] Motzer RJ, Bacik J, Murphy BA, Russo P, Mazumdar M.
Erratum in: Cancer 2006;107:2315. Interferon-alfa as a comparative treatment for clinical trials
[71] Peccatori J, Barkholt L, Demirer T, Sormani MP, Bruzzi P, of new therapies against advanced renal cell carcinoma. J
Ciceri F, et al. European Bone Marrow Transplantation Clin Oncol 2002;20:289–96.
Solid Tumor Working Party. Prognostic factors for survival [86] Motzer RJ, Bukowski RM, Figlin RA, Hutson TE,
in patients with advanced renal cell carcinoma undergoing Michaelson MD, Kim ST, et al. Prognostic nomogram for
nonmyeloablative allogeneic stem cell transplantation. Can- sunitinib in patients with metastatic renal cell carcinoma.
cer 2005;104:2099–103. Cancer 2008;113:1552–8.
[72] D’Amico AV, Chen MH, Cox MC, Dahut W, Figg WD. [87] Armitage JO, Weisenburger DD. New approach to classifying
Prostate-specific antigen response duration and risk of non-Hodgkin’s lymphomas: Clinical features of the major
death for patients with hormone-refractory metastatic pros- histologic subtypes. Non-Hodgkin’s Lymphoma Classifica-
tate cancer. Urology 2005;66:571–6. tion Project. J Clin Oncol 1998;16:2780–95.
[73] Taplin ME, George DJ, Halabi S, Sanford B, Febbo PG, [88] Scher HI, Heller G, Molina A, Attard G, Danila DC, Jia X,
Hennessy KT, et al. Prognostic significance of plasma et al. circulating tumor cell biomarker panel as an individual-
chromogranin a levels in patients with hormone-refractory level surrogate for survival in metastatic castration-
prostate cancer treated in Cancer and Leukemia Group B resistant prostate cancer. J Clin Oncol Epub 2015 Mar 23.
9480 study. Urology 2005;66:386–91. pii: JCO.2014.55.3487.
[74] Schmidt H, Bastholt L, Geertsen P, Christensen IJ, Larsen S, [89] Taverna G, Pedretti E, Di Caro G, Borroni EM, Marchesi F,
Gehl J, et al. Elevated neutrophil and monocyte counts in Grizzi F. Inflammation and prostate cancer: Friends or foe?
peripheral blood are associated with poor survival in patients Inflamm Res Epub 2015 Mar 19.
with metastatic melanoma: A prognostic model. Br J Cancer [90] Langsenlehner T, Pichler M, Thurner EM, Krenn-Pilko S,
2005;93:273–8. Stojakovic T, Gerger A, et al. Evaluation of the platelet-
[75] Soubrane C, Rixe O, Meric JB, Khayat D, Mouawad R. to-lymphocyte ratio as a prognostic indicator in a European
Pretreatment serum interleukin-6 concentration as a prog- cohort of patients with prostate cancer treated with
nostic factor of overall survival in metastatic malignant radiotherapy. Urol Oncol Epub 2015 Mar 10. pii: S1078–
melanoma patients treated with biochemotherapy: A retro- 1439(15)00062–9.
spective study. Melanoma Res 2005;15:199–204. [91] Graff JN, Beer TM, Liu B, Sonpavde G, Taioli E. Pooled
[76] George DJ, Halabi S, Shepard TF, Sanford B, Vogelzang NJ, analysis of C-reactive protein levels and mortality in pros-
Small EJ, et al. The prognostic significance of plasma tate cancer patients. Clin Genitourin Cancer Epub 2015
interleukin-6 levels in patients with metastatic hormone- Jan 30. pii: S1558–7673(15)00013–0.
refractory prostate cancer: Results from cancer and leukemia [92] van Soest RJ, Templeton AJ, Vera-Badillo FE, Mercier F,
group B 9480. Clin Cancer Res 2005;11:1815–20. Sonpavde G, Amir E, et al. Neutrophil-to-lymphocyte ratio
[77] Masaki T, Ohkawa S, Amano A, Ueno M, Miyakawa K, as a prognostic biomarker for men with metastatic castration-
Tarao K. Noninvasive assessment of tumor vascularity by resistant prostate cancer receiving first-line chemotherapy:
contrast-enhanced ultrasonography and the prognosis of Data from two randomized phase III trials. Ann Oncol
patients with nonresectable pancreatic carcinoma. Cancer 2015;26:743–9.
2005;103:1026–35. [93] Fizazi K, Massard C, Smith M, Rader M, Brown J, Milecki P,
[78] Ando S, Suzuki M, Yamamoto N, Iida T, Kimura H. The et al. Bone-related parameters are the main prognostic factors
prognostic value of both neuron-specific enolase (NSE) and for overall survival in men with bone metastases from castra-
Cyfra21-1 in small cell lung cancer. Anticancer Res tion-resistant prostate cancer. Eur Urol 2014 Oct 29. pii:
2004;24:1941–6. S0302–2838(14)01009–4.
[79] Chiarion-Sileni V, Del Bianco P, De Salvo GL, Lo Re G, [94] Armstrong AJ, Garrett-Mayer E, de Wit R, Tannock I,
Romanini A, Labianca R, et al. Italian Melanoma Inter- Eisenberger M. Prediction of survival following first-line
group (IMI). Quality of life evaluation in a randomised trial chemotherapy in men with castration-resistant metastatic
of chemotherapy versus bio-chemotherapy in advanced prostate cancer. Clin Cancer Res 2010;16:203–11.
melanoma patients. Eur J Cancer 2003;39:1577–85. [95] Denko NC. Hypoxia. HIF1 and glucose metabolism in the
[80] Atzpodien J, Royston P,Wandert T, Reitz M; DGCIN – German solid tumour. Nat Rev Cancer 2008;8:705–13.
Cooperative Renal Carcinoma Chemo-Immunotherapy Trials [96] Sonveaux P, Végran F, Schroeder T, Wergin MC, Verrax J,
Group. Metastatic renal carcinoma comprehensive prognostic Rabbani ZN, et al. Targeting lactate-fueled respiration
system. Br J Cancer 2003;88:348–53. selectively kills hypoxic tumor cells in mice. J Clin Invest
[81] Lagerwaard FJ, Levendag PC, Nowak PJ, Eijkenboom WM, 2008;118:3930–42.
Hanssens PE, Schmitz PI. Identification of prognostic factors [97] Feron O. Pyruvate into lactate and back: From the Warburg
in patients with brain metastases: A review of 1292 patients. effect to symbiotic energy fuel exchange in cancer cells.
Int J Radiat Oncol Biol Phys 1999;43:795–803. Radiother Oncol 2009;92:329–33.
[82] Franzke A, Probst-Kepper M, Buer J, Duensing S, [98] Whitaker-Menezes D, Martinez-Outschoorn UE, Lin Z,
Hoffmann R, Wittke F, et al. Elevated pretreatment serum Ertel A, Flomenberg N, Witkiewicz AK, et al. Evidence for
levels of soluble vascular cell adhesion molecule 1 and lactate a stromal-epithelial ‘lactate shuttle’ in human tumors:
dehydrogenase as predictors of survival in cutaneous meta- MCT4 is a marker of oxidative stress in cancer-associated
static malignant melanoma. Br J Cancer 1998;78:40–5. fibroblasts. Cell Cycle 2011;10:1772–83.
[83] Fosså SD, Oliver RT, Stenning SP, Horwich A, Wilkinson P, [99] Semenza GL, Jiang BH, Leung SW, Passantino R, Concor-
Read G, et al. Prognostic factors for patients with advanced det JP, Maire P, et al. Hypoxia response elements in the
seminoma treated with platinum-based chemotherapy. Eur aldolase A, enolase 1, and lactate dehydrogenase A gene
J Cancer 1997;33:1380–7. promoters contain essential binding sites for hypoxia-induc-
[84] van Dijk MR, Steyerberg EW, Habbema JD. Survival of ible factor 1. J Biol Chem 1996;271:32529–37.
non-seminomatous germ cell cancer patients according to [100] Faloppi L, Scartozzi M, Bianconi M, Svegliati Baroni G,
the IGCC classification: An update based on meta-analysis. Toniutto P, Giampieri R, et al. The role of LDH serum
Eur J Cancer 2006;42:820–6. levels in predicting global outcome in HCC patients treated
970 F. Petrelli et al.
with sorafenib: Implications for clinical management. BMC drogenase-A re-sensitizes taxol-resistant cancer cells to
Cancer 2014;14:110. taxol. Mol Cancer 2010;9:33.
[101] Scartozzi M, Giampieri R, Maccaroni E, Del Prete M, [106] Zhai X, Yang Y, Wan J, Zhu R, Wu Y. Inhibition of LDH-A
Faloppi L, Bianconi M, et al. Pre-treatment lactate by oxamate induces G2/M arrest, apoptosis and increases
dehydrogenase levels as predictor of efficacy of first-line radiosensitivity in nasopharyngeal carcinoma cells. Oncol
bevacizumab-based therapy in metastatic colorectal cancer Rep 2013;30:2983–91.
patients. Br J Cancer 2012;106:799–804. [107] Gupta GS. LDH-C4: A target with therapeutic potential for
[102] Xie H, Hanai J, Ren JG, Kats L, Burgess K, Bhargava P, cancer and contraception. Mol Cell Biochem 2012;371:
et al. Targeting lactate dehydrogenase – a inhibits tumori- 115–27.
genesis and tumor progression in mouse models of lung [108] Wang ZY, Loo TY, Shen JG, Wang N, Wang DM,Yang DP, et al.
cancer and impacts tumor-initiating cells. Cell Metab LDH-A silencing suppresses breast cancer tumorigenicity through
2014;19:795–809. induction of oxidative stress mediated mitochondrial pathway
[103] Liu X, Yang Z, Chen Z, Chen R, Zhao D, Zhou Y, et al. apoptosis. Breast Cancer Res Treat 2012;131:791–800.
Effects of the suppression of lactate dehydrogenase A on [109] Fantin VR, St-Pierre J, Leder P. Attenuation of LDH-A
the growth and invasion of human gastric cancer cells. expression uncovers a link between glycolysis, mitochon-
Oncol Rep 2015;33:157–62. drial physiology, and tumor maintenance. Cancer Cell
[104] Xie H, Valera VA, Merino MJ, Amato AM, Signoretti S, 2006;9:425–34. Erratum in: Cancer Cell 2006;10:172.
Linehan WM, et al. LDH-A inhibition, a therapeutic strategy [110] Granchi C, Roy S, Giacomelli C, Macchia M, Tuccinardi T,
for treatment of hereditary leiomyomatosis and renal cell Martinelli A, et al. Discovery of N-hydroxyindole-based inhibi-
cancer. Mol Cancer Ther 2009;8:626–35. tors of human lactate dehydrogenase isoform A (LDH-A) as
[105] Zhou M, Zhao Y, Ding Y, Liu H, Liu Z, Fodstad O, et al. starvation agents against cancer cells. J Med Chem 2011;54:
Warburg effect in chemosensitivity: Targeting lactate dehy- 1599–612.

Supplementary material available online


Supplementary Figure 1 and Table I available online
at http://informahealthcare.com/doi/abs/10.3109/
0284186X.2015.1043026.

You might also like