Open Access

High mitochondrial content is associated with breast cancer aggressiveness

  • Authors:
    • Patrick Lebok
    • Katharina Schütt
    • Martina Kluth
    • Isabell Witzel
    • Linn Wölber
    • Peter Paluchowski
    • Luigi Terracciano
    • Christian Wilke
    • Uwe Heilenkötter
    • Volkmar Müller
    • Barbara Schmalfeldt
    • Ronald Simon
    • Guido Sauter
    • Ingo Von Leffern
    • Till Krech
    • Rainer Horst Krech
    • Frank Jacobsen
    • Eike Burandt
  • View Affiliations

  • Published online on: August 8, 2021     https://doi.org/10.3892/mco.2021.2365
  • Article Number: 203
  • Copyright: © Lebok et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Mitochondria are relevant for cancer initiation and progression. Antibodies against mitochondrially encoded cytochrome c oxidase II (MTCO2), targeting a mitochondria specific epitope, can be used to quantitate the mitochondria content of tumor cells. The present study evaluated the impact of the cellular mitochondrial content on the prognosis of patients with breast cancer using immunohistochemical analysis on 2,197 arrayed breast cancer specimens. Results were compared with histological tumor parameters, patient overall survival, tumor cell proliferation using Ki67 labeling index (Ki67LI) and various other molecular features. Tumor cells exhibited stronger MTCO2 expression than normal breast epithelial cells. MTCO2 immunostaining was largely absent in normal breast epithelium, but was observed in 71.9% of 1,797 analyzable cancer specimens, including 34.6% tumors with weak expression, 22.3% with moderate expression and 15.0% with strong expression. High MTCO2 expression was significantly associated with advanced tumor stage, high Bloom‑Richardson‑Elston/Nottingham (BRE) grade, nodal metastasis and shorter overall survival (P<0.0001 each). In multivariate analysis, MTCO2 expression did not provide prognostic information independent of BRE grade, pathological tumor and pathological lymph node status. Additionally, significant associations were observed for high MTCO2 expression and various molecular features, including high Ki67LI, amplifications of HER2, MYC, CCND1 and MDM2, deletions of PTEN, 8p21 and 9p, low estrogen receptor expression (P<0.0001 each) and progesterone receptor expression (P<0.0001). The present study demonstrated that high MTCO2 expression was strongly associated with a poor prognosis and unfavorable phenotypical and molecular tumor features in patients with breast cancer. This suggests that the mitochondrial content may have a pivotal role in breast cancer progression.

Introduction

Breast cancer, the most common malignancy in women (1), is treated by surgical removal of the cancer. In addition, adjuvant systemic therapy is given depending on the perceived aggressiveness of the removed cancer. Currently the established prognostic parameter include histological grade, tumor size, presence of lymph node metastasis, tumor cell proliferation (Ki67 labeling index; Ki67LI) as well as hormonal receptor and HER2 status (2-4) (Ki67) (5). In many patients, supplementary molecular parameters are analyzed (6-8). These molecular classifiers are built on multiplexed analysis of the mRNAs of 21-70 genes (9-11).

The rising interest in mitochondrial function and dysfunction on cancer development has been reviewed by Davis and Williams and Hsu et al (12,13). The loss of proliferation control in cancer cells may result in cellular masses that extend beyond the capacity of the supporting vasculature, leading to oxygen and nutrient deprivation. Hence, tumor cells must adapt to overcome these restrictions. Mitochondria are key organelles for energy production in normal and neoplastic cells. Quantity and activity of mitochondria are essential for tumor growth (reviewed in refs. 12-16). Mutations in mitochondrial genes or aberrant mitochondrial content have been described to occur in various cancer types (17-20). An increased mitochondria quantity has earlier been linked to aggressive tumor phenotype and poor prognosis in lung (21), colorectal (22,23), prostate (24), gastric (25), cervical (18), and ovarian cancer (26). In glioma, however, high mitochondria content was linked to favorable prognosis (27). In one study, on 76 breast carcinomas, a prognostic impact of the mitochondria count was also suggested (28). Focused on these reports, we assumed that the cellular mitochondria content of breast cancer cells might potentially be clinically relevant in breast cancer.

The mitochondrially encoded cytochrome c oxidase II (MTCO2) monoclonal antibody recognizes a 60 kDa non-glycosylated protein subunit of cytochrome c oxidase in mitochondria found in human cells and has been used to reveal the mitochondrial content of tumor cells in previous studies (24,28,29). We tested the clinical relevance of the cellular mitochondria content in breast cancer on a pre-existing breast cancer tissue microarray (TMA) containing more than 2,000 cancers. The data show that a ʻmitochondrion-rich phenotype’ represents a strong and independent predictor of patient prognosis in breast cancer.

Materials and methods

Patients

A total of 2,197 human breast cancer samples from paraffin-embedded tissue specimens fixed in 4% neutral buffered formalin were used (30). The breast cancer samples were consecutively collected between 1984 and 2000 and follow-up data were retrospectively collected. The median patient's age was 63 (range, 25-101) years. Overall survival data were available from 1,982 patients (713 patients with and 1,508 without event). The mean follow-up time was 63 months (range, 1-176 months). The TMA was produced as detailed earlier in (31). In short, from each patient one 0.6 mm core was taken from a representative cancer tissue block. All tissues were distributed among 6 TMA blocks, each containing 263-522 tumor samples. Four-micrometre sections of the TMA blocks were transferred to an adhesive coated slide system (Instrumedics Inc.) for immunohistochemistry (IHC) analysis. Molecular data used in this study were available from previously published studies. These included amplification/deletion data obtained by fluorescence in situ hybridization for HER2, MYC, 8p21, 9p21, and PTEN, as well as Ki67LI (30,32-34).

IHC

Freshly cut TMA sections were processed the same day. Slides were deparaffinized and exposed to heat-induced antigen retrieval for 5 min at 121˚C in pH 7.8 Tris-EDTA-Citrate buffer prior to incubation with the mouse monoclonal antibody MTCO2 (Abcam; #ab3298; 1/450 dilution). Bound antibody was visualized using the EnVision kit (Dako). MTCO2 staining was homogenous in the analyzed tissue samples and staining intensity of all cases was semiquantitatively assessed in four categories: Negative (no visible staining), weak (1+ staining intensity), moderate (2+ stainong intensity) and strong (3+ staining intensity).

Statistical analysis

Contingency tables were calculated to study associations between MTCO2 expression and clinicopathological variables, and the chi-square (likelihood) test was used to find significant relationships. Analysis of variance and F-test was applied to find associations between MTCO2 staining levels and tumor cell proliferation as measured by the Ki67LI. Kaplan-Meier curves were generated using overall survival as the clinical endpoint. The log-rank test was applied to test the significance of differences between stratified survival functions. Cox proportional hazards regression analysis was performed to test the statistical independence and significance between pathological and molecular variables. JMP 12.0 software (SAS Institute Inc.) was used.

Results

Technical issues

A total of 1,797 (81.8%) of the 2,197 arrayed tumor samples were interpretable in our TMA analysis. Non-informative cases (400 spots; 18.2%) were due to missing tissue samples or the absence of unequivocal cancer tissue in the TMA spot.

MTCO2 immunostaining in normal breast tissue and breast cancer

There were 20 normal breast tissue samples included in our TMA. Normal breast tissues showed negative to moderate MTCO2 staining in luminal cells under the chosen experimental conditions. In cancer, MTCO2 immunostaining was considered weak in 34.6%, moderate in 22.3% and strong in 15.0% of tumors. A total of 506 (28.2%) showed no detectable MTCO2 staining and were categorized as negative. Characteristic images of MTCO2 immunostainings are shown in Fig. 1. The intensity of MTCO2 immunostaining varied between histological breast cancer subtypes (Table I). Strong MTCO2 staining was significantly more common in medullary (27.9%), papillary (16.0%) and cancers of no special type (NST; 16.6%) than in lobular (6.9%) or tubular carcinomas (4.9%). Strong MTCO2 staining was also commonly seen in some of the rare breast cancer subtypes such as in 3 of 13 carcinomas with apocrine differentiation, 17 of 61 carcinomas with medullary features and 2 of 12 glycogen-rich clear cell type carcinomas (Table SI).

Table I

Association between MTCO2 staining and breast cancer phenotype.

Table I

Association between MTCO2 staining and breast cancer phenotype.

 MTCO2 staining 
CharacteristicsNNegative, %Weak, %Moderate, %Strong, %P-value
All cases1,79728.234.622.315.0 
Histology      
     NST1,28124.236.023.216.6 
     Lobular carcinoma23346.433.113.76.9 <0.0001a
     Medullary carcinoma6118.026.227.927.90.0761a
     Cribriform carcinoma5534.627.321.816.40.3461a
     Mucinous carcinoma5145.133.319.62.00.0005a
     Tubular carcinoma4148.839.07.34.90.0004a
     Papillary carcinoma2520.028.036.016.00.5425a
     Apocrine carcinoma1323.115.438.523.10.3460a
     Other rare typesb229.127.350.013.60.0399a
pT stage      
     pT163136.340.317.65.9<0.0001 (<0.0001c)
     pT285124.232.124.719.0 
     pT39825.530.624.519.4 
     pT420921.130.124.923.9 
BRE grade      
     G142341.637.813.57.1<0.0001 (<0.0001c)
     G267329.935.523.511.1 
     G356415.426.629.628.4 
Nodal stage      
     pN076133.534.222.79.6<0.0001 (0.0063c)
     pN164425.636.220.317.9 
     pN210318.538.824.318.5 
     pN300.00.00.00.0 
Estrogen receptor      
     Negative40616.0129.3128.8225.86<0.0001 (<0.0001c)
     Positive1,29631.3336.520.4511.73 
Progesterone receptor      
     Negative1,05929.2732.0122.9515.77<0.0001 (0.0795c)
     Positive56926.1940.0719.6814.06 

[i] avs. NST;

[ii] bOther types included adenoid-cystic carcinoma, carcinoma with apocrine differentiation, carcinoma with medullary features, carcinoma with neuroendocrine features, carcinoma with signet-ring-cell differentiation, glycogen-rich clear cell carcinoma, metaplastic carcinoma of NST and lipid-rich carcinoma;

[iii] cin NST only. Numbers do not always add up to 1,797 in the different categories due to cases with missing data. NST, no special type; pT, pathological tumor; pN, pathological lymph node; MTCO2, mitochondrially encoded cytochrome c oxidase II; BRE, Bloom-Richardson-Elston/Nottingham system.

Association with tumor phenotype and molecular features

High levels of MTCO2 immunostaining were significantly associated with high pT stage, high BRE grade, estrogen and progesterone receptor negativity as well as HER2 overexpression or amplification (P<0.0001 each, Tables I and II). This was also seen for NST carcinomas (P≤0.01, Table I). Further analyses with previously described frequent and prognostic relevant molecular features of breast cancers such as HER2(35), and c-MYC- amplification (32) as well as deletions of 8p21(34), 9p21(33), and 10q23(36) showed significant associations with high MTCO2 staining intensity (Table II).

Table II

Association between MTCO2 staining and molecular alterations.

Table II

Association between MTCO2 staining and molecular alterations.

 MTCO2 staining 
Molecular alterationsNNegative, %Weak, %Moderate, %Strong, %P-value
HER2 normal1,14129.236.321.213.3<0.0001
HER2 amplified23915.532.630.121.8 
MYC normal1,23226.934.923.115.1<0.0001
MYC amplified647.829.728.134.4 
8p21 normal57827.739.120.912.3<0.0001
8p21 deletion55317.027.729.525.9 
9p21 normal83525.033.124.417.50.0182
9p21 deletion15016.728.032.023.3 
10q23 normal90425.035.022.917.1<0.0001
10q23 deletion21611.626.436.125.9 

[i] MTCO2, mitochondrially encoded cytochrome c oxidase II.

Association with tumor cell proliferation

Data on tumor cell proliferation as evaluated by the Ki67LI were available from a previous study with the same TMA (30). The mean Ki67LI increased from 19.62±0.66 in MTCO2 negative cancers to 37.75±0.93 in cancers with strong MTCO2 staining (P<0.0001). This statistically significant relationship was also seen in tumor subsets with identical pT or pN stage, lobular and medullary carcinoma, BRE grade and HER2 status as well as 8p and PTEN deletion. All data are summarized in Table III.

Table III

Association between MTCO2 staining and Ki67LI.

Table III

Association between MTCO2 staining and Ki67LI.

CasesMTCO2 stainingNKi67LIP-value
All casesNegative42819.6±0.7<0.0001
 Weak52327.0±0.6 
 Moderate33833.0±0.8 
 Strong21637.8±0.9 
No special typeNegative26420.7±0.8<0.0001
 Weak38327.8±0.7 
 Moderate25333.3±0.9 
 Strong16838.0±1.1 
Lobular cancerNegative9216.2±1.2<0.0001
 Weak6620.3±1.4 
 Moderate2428.4±2.3 
 Strong1526.9±2.9 
Medullary cancerNegative929.9±5.20.0109
 Weak1543.7±4.1 
 Moderate1650.2±3.9 
 Strong1550.9±4.0 
HER2 amplifiedNegative3226.7±2.3<0.0001
 Weak6734.2±1.6 
 Moderate6440.3±1.6 
 Strong4341.3±1.9 
MYC amplifiedNegative428.5±7.40.3927
 Weak1938.3±3.4 
 Moderate1741.6±3.6 
 Strong2141.6±3.2 
8p deletionNegative8624.8±1.5<0.0001
 Weak13530.2±1.2 
 Moderate14535.3±1.2 
 Strong11640.3±1.3 
PTEN deletionNegative2430.6±3.20.0118
 Weak5537.7±2.1 
 Moderate7541.7±1.8 
 Strong4442.2±2.3 
pT1Negative19219.0±0.9<0.0001
 Weak20023.8±0.9 
 Moderate9029.9±1.3 
 Strong3137.8±2.3 
pT2Negative17019.9±1.1<0.0001
 Weak23829.6±0.9 
 Moderate17935.3±1.1 
 Strong12737.9±1.3 
pT3Negative2318.2±3.1<0.0001
 Weak2731.2±2.9 
 Moderate2230.1±3.2 
 Strong1643.8±3.7 
pT4Negative4121.9±2.2<0.0001
 Weak5725.3±1.7 
 Moderate4431.9±1.9 
 Strong4134.9±2.1 
BRE G1Negative15015.5±0.8<0.0001
 Weak12719.5±0.9 
 Moderate4521.4±1.5 
 Strong2526.4±1.9 
BRE G2Negative17018.8±0.9<0.0001
 Weak20823.7±0.8 
 Moderate13428.9±01.0 
 Strong6331.4±1.4 
BRE G3Negative7629.4±1.7<0.0001
 Weak13037.5±1.3 
 Moderate14540.4±1.2 
 Strong12443.2±1.3 
pN0Negative21919.5±0.9<0.0001
 Weak21626.5±0.9 
 Moderate14434.5±1.1 
 Strong5938.9±1.8 
pN1Negative13819.4±1.2<0.0001
 Weak19827.3±1.0 
 Moderate11132.2±1.3 
 Strong9239.1±1.4 
pN2Negative1825.6±3.20.0064
 Weak3429.4±2.3 
 Moderate2333.4±2.8 
 Strong1641.4±3.4 

[i] Ki67LI, Ki67 labeling index; pT, pathological tumor; pN, pathological lymph node; MTCO2, mitochondrially encoded cytochrome c oxidase II; G, grade; BRE, Bloom-Richardson-Elston/Nottingham system.

Prognostic significance of MTCO2 expression

Survival data were available for 1,806 cancers with interpretable IHC results. The rate of surviving patients continuously decreased with increasing levels of MTCO2 immunostaining (P=0.0001; Fig. 2). The association between strong MTCO2 immunostaining and poor prognosis was also seen in the subgroup of NST cancers (P<0.0001; Fig. 2) and in the nodal positive subset (P<0.0001; Fig. 2) and to a much lesser extent also in nodal negative NST cancers (P=0.0418; Fig. 2). Multivariate analysis for NST cancers including pT stage, nodal status, and BRE grade did not identify MTCO2 immunostaining as an independent prognosticator of survival, however (Table IV).

Table IV

Multivariate analysis in all breast cancer cases (n=1,377).

Table IV

Multivariate analysis in all breast cancer cases (n=1,377).

CharacteristicsHazard ratioP-valueOverall P-value
pT stage   
     2 vs. 11.520.0010<0.0001
     3 vs. 21.050.7700 
     4 vs. 21.650.0006 
     4 vs. 31.560.2230 
BRE grade  <0.0001
     G2 vs. G11.350.0522 
     G3 vs. G12.81<0.0001 
     G3 vs. G22.08<0.0001 
pN  <0.0001
     1 vs. 02.26<0.0001 
     2 vs. 12.33<0.0001 
     2 vs. 05.27<0.0001 
MTCO2 staining  0.1464
     Weak vs. negative1.330.0396 
     Moderate vs. weak1.010.9109 
     Strong vs. moderate0.880.4133 

[i] pT, pathological tumor; pN, pathological lymph node; MTCO2, mitochondrially encoded cytochrome c oxidase II; G, grade; BRE, Bloom-Richardson-Elston/Nottingham system.

Discussion

Our study shows that high mitochondria content is significantly linked to disadvantageous tumor phenotype and bad prognosis in breast cancer.

MTCO2 immunostaining is highly specific for the mitochondrial DNA encoded second subunit of cytochrome c oxidase and can thus be used to quantitate the mitochondria content by IHC (29). Although mitochondria are present in every normal and neoplastic human cell, 28.2% of our tumors had a negative staining result. This was due to our approach to define experimental conditions, which distinguish cancers with low and high mitochondria quantities. The higher level of MTCO2 immunostaining in breast cancers as compared to normal breast tissues fits with the concept that neoplastic transformation goes along with higher cellular activity requiring more active mitochondria. That a striking further increase of MTCO2 immunostaining was detected with rising tumor grade and stage, demonstrates that elevated numbers of mitochondria are also supporting cancer progression. This is consistent with increasing energy requirement and a rearranged metabolism during tumor progression. Our data fit well with findings in multiple other cancer types, including lung (21), colorectal (22,23), prostate (24), gastric (25), cervical (18), and ovarian cancer (26), where a similar link between high levels of MTCO2 with adverse tumor phenotype and bad prognosis was shown.

In this study, a ubiquitously expressed protein was quantitated by IHC. The TMA approach is optimal for the identification of subtle staining differences of proteins that are abundantly present in cancer, such as mitochondrial components, because TMAs enable maximal experimental standardization at all levels. In our study, more than 1,700 breast cancers were analyzed the same day for maximal standardization. Moreover, all TMA sections were cut on one day immediately before staining in order to avoid unequal decay of a tissues reactivity to antibody binding (37). Finally, one pathologist interpreted all immunostainings in one continuous session to enable maximal standardization of staining interpretation. In earlier studies, this breast cancer TMA enabled us to validate the prognostic impact of several well-established prognostic biomarkers, such as HER2 alterations, estrogen and progesterone receptor expression (30), high Ki67LI, nuclear p53 accumulation (30), and PTEN deletion (34). These earlier data demonstrate the utility of our patient cohort to identify prognostic biomarkers.

The molecular database that has been collected during earlier studies for our set of cancers offers the advantage that biomarkers of interest can always be compared with preexisting data. For the purpose of this study, we had selected HER2 amplification as well as estrogen and progesterone receptor expression because of their central role in breast cancer. The strong link between MTCO2 expression and these important features further illustrates the importance of the mitochondria quantity in breast cancer. Our analyses also included Ki67LI as another pivotal parameter for cellular activity and various further chromosomal deletions and amplifications because of the role of some of them for regulating mitochondrion homeostasis.

Mitochondrial homeostasis is critical for cancer. A sufficiently high production of mitochondria is required to suffice the needs for energy production and cell metabolism. The prominent association found between c-Myc amplification and high MTCO2 expression fits well with the key role of c-Myc as an activator of mitochondrial biogenesis in cancer (38-40). The transcription factor c-Myc is best known for its critical role in cell cycle regulation, cell growth, metabolism and apoptosis (41-43). However, c-Myc also targets more than 400 different mitochondrial genes (38-41,44). Studies have demonstrated that an elevated or reduced c-Myc protein quantity leads to an increased/diminished mitochondrial mass (45,46). This couples c-Myc's role of a key activator of cell cycle activity with mitochondrial biogenesis. As such, c-Myc increases cellular biosynthetic and respiratory capacity by upregulating mitochondrial metabolism to complement its effects on stimulating cell cycle progression to coordinate rapid cell growth (45,47).

A critical role of high mitochondrion count for cell proliferation in breast cancer is supported by our data showing a striking link between MTCO2 expression and a high Ki67LI which was also visible in the vast majority of groups defined by identical morphological or molecular features.

The PTEN-induced putative kinase 1 (PINK1)/Parkin pathway is a major inducer of mitophagy. It is triggered by mitochondrial membrane depolarization, a signal of mitochondrial dysfunction that results from lack of reducing equivalents, hypoxia and impaired electron transport [reviewed in (48)]. The conspicuous relationship between PTEN deletion and high MTCO2 staining in our study may thus indicate that high mitochondria quantities may also be caused by reduced mitophagy. Although clearance of damaged mitochondria via mitophagy is viewed to be also critical for cellular fitness since dysfunctional mitochondria can impair the electron transport chain function, reduced mitophagy can also promote cancer reviewed in ref. 49). Mitophagy-deficient Parkin null mice develop spontaneous hepatic tumors (50). Decreased mitophagy may allow for a permissive threshold of dysfunctional mitochondria to persist, generating increased tumor-promoting free oxygen radicals reviewed in ref. 49).

Cytochrome oxidase subunit 2 is a key enzyme of the respiratory chain, catalyzing electron transfer from NADH and succinate to molecular oxygen (51). It has no direct tumor related function but serves as a marker for the cellular mitochondria content. Increased mitochondria content in cancer cells often occurs as a result of the elevated metabolism and energy needs of expanding tumor cell populations (52). Although the mitochondrial content provided no additional prognostic information in multivariate analysis, the marked prognostic relevance of MTCO2 immunostaining found in this study may still suggest ʻmitochondria content’ as a biomarker with potential clinical utility. Molecular analyses are frequently done in breast cancer to better assess patient prognosis and to determine whether adjuvant chemotherapy should be applied (6-8). Most currently used tests are analyzing RNAs of multiple genes forming a prognostic score (9-11,53). RNA based tests share the disadvantage, however, that the analyzed RNA always represents a mixture of cancer cells and a variable fraction of non-neoplastic inflammatory and stromal cells. Now that multiplex fluorescent-based quantitative IHC becomes increasingly available, it is well possible that RNA based test will sooner or later be replaced by IHC based multi-gene tests. MTCO2 might be a candidate for being part of such a test, also because of the general biologic importance of mitochondria, which are also the target of several anti-cancer drugs under development reviewed in refs. 54-57).

It is a limitation of our study that MTCO2 IHC data highlight relevant associations between cancer phenotype and genotype but do not provide mechanistic insights into the putative cancer biological role of MTCO2. Further studies on the tumor relevant aspects of mitochondrial density and MTCO2 protein function are required to better understand the prognostic role of MTCO2 in breast cancer.

In summary, our findings identify MTCO2 immunostaining as a powerful prognostic biomarker in breast cancer. MTCO2 measurement, most likely in combination with other antibodies might be of clinical utility in breast cancer prognosis assessment.

Supplementary Material

MTC02 staining in rare breast cancer subtypes.

Acknowledgements

The authors would like to thank Ms. Inge Brandt and Ms. Sünje Seekamp from the Institute of Pathology of University Medical Center Hamburg-Eppendorf (Hamburg, Germany) for excellent technical assistance.

Funding

No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

All authors contributed to the conception and design of the study. PL, KS, MK, IW, LW, PP, LT, CW, UH, VM, BS, IvL, TK, RHK and FJ prepared the material, and collected and analyzed the data. PL, EB, RS, MK and GS wrote the first draft of the manuscript, and all authors commented on previous versions of the manuscript. RS, MK and GS confirmed the authenticity of all the raw data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The usage of archived diagnostic leftover tissues for manufacturing the tissue microarrays and their analysis for research purposes, as well as patient data analysis, has been approved by local laws (HmbKHG, §12) and by the local ethics committee (Ethics Commission of the Ärtzekammer Hamburg, Hamburg, Germany; approval no. WF-049/09). Informed consent was waived by the ethics committee due to the retrospective nature of the study. All work has been carried out in compliance with the Declaration of Helsinki.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2020. CA Cancer J Clin. 70:7–30. 2020.PubMed/NCBI View Article : Google Scholar

2 

Leong AS and Raymond WA: Prognostic parameters in breast cancer. Pathology. 21:169–175. 1989.PubMed/NCBI View Article : Google Scholar

3 

Taneja P, Maglic D, Kai F, Zhu S, Kendig RD, Fry EA and Inoue K: Classical and novel prognostic markers for breast cancer and their clinical significance. Clin Med Insights Oncol. 4:15–34. 2010.PubMed/NCBI View Article : Google Scholar

4 

Soliman NA and Yussif SM: Ki-67 as a prognostic marker according to breast cancer molecular subtype. Cancer Biol Med. 13:496–504. 2016.PubMed/NCBI View Article : Google Scholar

5 

Cao SS and Lu CT: Recent perspectives of breast cancer prognosis and predictive factors. Oncol Lett. 12:3674–3678. 2016.PubMed/NCBI View Article : Google Scholar

6 

Giuliano AE, Hunt KK, Ballman KV, Beitsch PD, Whitworth PW, Blumencranz PW, Leitch AM, Saha S, McCall LM and Morrow M: Axillary dissection vs no axillary dissection in women with invasive breast cancer and sentinel node metastasis: A randomized clinical trial. JAMA. 305:569–575. 2011.PubMed/NCBI View Article : Google Scholar

7 

McVeigh TP, Hughes LM, Miller N, Sheehan M, Keane M, Sweeney KJ and Kerin MJ: The impact of Oncotype DX testing on breast cancer management and chemotherapy prescribing patterns in a tertiary referral centre. Eur J Cancer. 50:2763–2770. 2014.PubMed/NCBI View Article : Google Scholar

8 

Naoi Y and Noguchi S: Multi-gene classifiers for prediction of recurrence in breast cancer patients. Breast Cancer. 23:12–18. 2016.PubMed/NCBI View Article : Google Scholar

9 

Hornberger J, Cosler LE and Lyman GH: Economic analysis of targeting chemotherapy using a 21-gene RT-PCR assay in lymph-node-negative, estrogen-receptor-positive, early-stage breast cancer. Am J Manag Care. 11:313–324. 2005.PubMed/NCBI

10 

Cobleigh MA, Tabesh B, Bitterman P, Baker J, Cronin M, Liu ML, Borchik R, Mosquera JM, Walker MG and Shak S: Tumor gene expression and prognosis in breast cancer patients with 10 or more positive lymph nodes. Clin Cancer Res. 11:8623–8631. 2005.PubMed/NCBI View Article : Google Scholar

11 

van 't Veer LJ, Dai H, van de Vijver MJ, He YD, Hart AA, Mao M, Peterse HL, van der Kooy K, Marton MJ, Witteveen AT, et al: Gene expression profiling predicts clinical outcome of breast cancer. Nature. 415:530–536. 2002.PubMed/NCBI View Article : Google Scholar

12 

Davis RE and Williams M: Mitochondrial function and dysfunction: An update. J Pharmacol Exp Ther. 342:598–607. 2012.PubMed/NCBI View Article : Google Scholar

13 

Hsu CC, Tseng LM and Lee HC: Role of mitochondrial dysfunction in cancer progression. Exp Biol Med (Maywood). 241:1281–1295. 2016.PubMed/NCBI View Article : Google Scholar

14 

Boland ML, Chourasia AH and Macleod KF: Mitochondrial dysfunction in cancer. Front Oncol. 3(292)2013.PubMed/NCBI View Article : Google Scholar

15 

Porporato PE, Filigheddu N, Pedro JMB, Kroemer G and Galluzzi L: Mitochondrial metabolism and cancer. Cell Res. 28:265–280. 2018.PubMed/NCBI View Article : Google Scholar

16 

Grasso D, Zampieri LX, Capeloa T, Van de Velde JA and Sonveaux P: Mitochondria in cancer. Cell Stress. 4:114–146. 2020.PubMed/NCBI View Article : Google Scholar

17 

Abu-Amero KK, Alzahrani AS, Zou M and Shi Y: High frequency of somatic mitochondrial DNA mutations in human thyroid carcinomas and complex I respiratory defect in thyroid cancer cell lines. Oncogene. 24:1455–1460. 2005.PubMed/NCBI View Article : Google Scholar

18 

Warowicka A, Kwasniewska A and Gozdzicka-Jozefiak A: Alterations in mtDNA: A qualitative and quantitative study associated with cervical cancer development. Gynecol Oncol. 129:193–198. 2013.PubMed/NCBI View Article : Google Scholar

19 

Gao JY, Song BR, Peng JJ and Lu YM: Correlation between mitochondrial TRAP-1 expression and lymph node metastasis in colorectal cancer. World J Gastroenterol. 18:5965–5971. 2012.PubMed/NCBI View Article : Google Scholar

20 

Qian XL, Li YQ, Gu F, Liu FF, Li WD, Zhang XM and Fu L: Overexpression of ubiquitous mitochondrial creatine kinase (uMtCK) accelerates tumor growth by inhibiting apoptosis of breast cancer cells and is associated with a poor prognosis in breast cancer patients. Biochem Biophys Res Commun. 427:60–66. 2012.PubMed/NCBI View Article : Google Scholar

21 

Sotgia F and Lisanti MP: Mitochondrial markers predict survival and progression in non-small cell lung cancer (NSCLC) patients: Use as companion diagnostics. Oncotarget. 8:68095–68107. 2017.PubMed/NCBI View Article : Google Scholar

22 

Ambrosini-Spaltro A, Salvi F, Betts CM, Frezza GP, Piemontese A, Del Prete P, Baldoni C, Foschini MP and Viale G: Oncocytic modifications in rectal adenocarcinomas after radio and chemotherapy. Virchows Arch. 448:442–448. 2006.PubMed/NCBI View Article : Google Scholar

23 

Wang Y, He S, Zhu X, Qiao W and Zhang J: High copy number of mitochondrial DNA predicts poor prognosis in patients with advanced stage colon cancer. Int J Biol Markers. 31:e382–e388. 2016.PubMed/NCBI View Article : Google Scholar

24 

Grupp K, Jedrzejewska K, Tsourlakis MC, Koop C, Wilczak W, Adam M, Quaas A, Sauter G, Simon R, Izbicki JR, et al: High mitochondria content is associated with prostate cancer disease progression. Mol Cancer. 12(145)2013.PubMed/NCBI View Article : Google Scholar

25 

Sotgia F and Lisanti MP: Mitochondrial biomarkers predict tumor progression and poor overall survival in gastric cancers: Companion diagnostics for personalized medicine. Oncotarget. 8:67117–67128. 2017.PubMed/NCBI View Article : Google Scholar

26 

Hu B and Guo Y: Inhibition of mitochondrial translation as a therapeutic strategy for human ovarian cancer to overcome chemoresistance. Biochem Biophys Res Commun. 509:373–378. 2019.PubMed/NCBI View Article : Google Scholar

27 

Zhang Y, Qu Y, Gao K, Yang Q, Shi B, Hou P and Ji M: High copy number of mitochondrial DNA (mtDNA) predicts good prognosis in glioma patients. Am J Cancer Res. 5:1207–1216. 2015.PubMed/NCBI

28 

Ragazzi M, de Biase D, Betts CM, Farnedi A, Ramadan SS, Tallini G, Reis-Filho JS and Eusebi V: Oncocytic carcinoma of the breast: Frequency, morphology and follow-up. Hum Pathol. 42:166–175. 2011.PubMed/NCBI View Article : Google Scholar

29 

Williams SL, Valnot I, Rustin P and Taanman JW: Cytochrome c oxidase subassemblies in fibroblast cultures from patients carrying mutations in COX10, SCO1, or SURF1. J Biol Chem. 279:7462–7469. 2004.PubMed/NCBI View Article : Google Scholar

30 

Ruiz C, Seibt S, Al Kuraya K, Siraj AK, Mirlacher M, Schraml P, Maurer R, Spichtin H, Torhorst J, Popovska S, et al: Tissue microarrays for comparing molecular features with proliferation activity in breast cancer. Int J Cancer. 118:2190–2194. 2006.PubMed/NCBI View Article : Google Scholar

31 

Mirlacher M and Simon R: Recipient block TMA technique. Methods Mol Biol. 664:37–44. 2010.PubMed/NCBI View Article : Google Scholar

32 

Al-Kuraya K, Schraml P, Torhorst J, Tapia C, Zaharieva B, Novotny H, Spichtin H, Maurer R, Mirlacher M, Köchli O, et al: Prognostic relevance of gene amplifications and coamplifications in breast cancer. Cancer Res. 64:8534–8540. 2004.PubMed/NCBI View Article : Google Scholar

33 

Lebok P, Roming M, Kluth M, Koop C, Özden C, Taskin B, Hussein K, Lebeau A, Witzel I, Wölber L, et al: p16 overexpression and 9p21 deletion are linked to unfavorable tumor phenotype in breast cancer. Oncotarget. 7:81322–81331. 2016.PubMed/NCBI View Article : Google Scholar

34 

Lebok P, Mittenzwei A, Kluth M, Özden C, Taskin B, Hussein K, Möller K, Hartmann A, Lebeau A, Witzel I, et al: 8p deletion is strongly linked to poor prognosis in breast cancer. Cancer Biol Ther. 16:1080–1087. 2015.PubMed/NCBI View Article : Google Scholar

35 

Yan M, Schwaederle M, Arguello D, Millis SZ, Gatalica Z and Kurzrock R: HER2 expression status in diverse cancers: Review of results from 37,992 patients. Cancer Metastasis Rev. 34:157–164. 2015.PubMed/NCBI View Article : Google Scholar

36 

Lebok P, Kopperschmidt V, Kluth M, Hube-Magg C, Özden C, B T, Hussein K, Mittenzwei A, Lebeau A, Witzel I, et al: Partial PTEN deletion is linked to poor prognosis in breast cancer. BMC Cancer. 15:963–910. 2015.PubMed/NCBI View Article : Google Scholar

37 

Simon R, Mirlacher M and Sauter G: Immunohistochemical analysis of tissue microarrays. Methods Mol Biol. 664:113–126. 2010.PubMed/NCBI View Article : Google Scholar

38 

Nieminen AI, Partanen JI, Hau A and Klefstrom J: c-Myc primed mitochondria determine cellular sensitivity to TRAIL-induced apoptosis. EMBO J. 26:1055–1067. 2007.PubMed/NCBI View Article : Google Scholar

39 

Desbiens KM, Deschesnes RG, Labrie MM, Desfossés Y, Lambert H, Landry J and Bellmann K: c-Myc potentiates the mitochondrial pathway of apoptosis by acting upstream of apoptosis signal-regulating kinase 1 (Ask1) in the p38 signalling cascade. Biochem J. 372:631–641. 2003.PubMed/NCBI View Article : Google Scholar

40 

Klefstrom J, Verschuren E and Evan G: c-Myc augments the apoptotic activity of cytosolic death receptor signaling proteins by engaging the mitochondrial apoptotic pathway. J Biol Chem. 277:43224–43232. 2002.PubMed/NCBI View Article : Google Scholar

41 

Dang CV, O'Donnell KA, Zeller KI, Nguyen T, Osthus RC and Li F: The c-Myc target gene network. Semin Cancer Biol. 16:253–264. 2006.PubMed/NCBI View Article : Google Scholar

42 

Amati B, Frank SR, Donjerkovic D and Taubert S: Function of the c-Myc oncoprotein in chromatin remodeling and transcription. Biochim Biophys Acta. 1471:M135–M145. 2001.PubMed/NCBI View Article : Google Scholar

43 

Dang CV, Resar LM, Emison E, Kim S, Li Q, Prescott JE, Wonsey D and Zeller K: Function of the c-Myc oncogenic transcription factor. Exp Cell Res. 253:63–77. 1999.PubMed/NCBI View Article : Google Scholar

44 

Wonsey DR, Zeller KI and Dang CV: The c-Myc target gene PRDX3 is required for mitochondrial homeostasis and neoplastic transformation. Proc Natl Acad Sci USA. 99:6649–6654. 2002.PubMed/NCBI View Article : Google Scholar

45 

Li F, Wang Y, Zeller KI, Potter JJ, Wonsey DR, O'Donnell KA, Kim JW, Yustein JT, Lee LA and Dang CV: Myc stimulates nuclearly encoded mitochondrial genes and mitochondrial biogenesis. Mol Cell Biol. 25:6225–6234. 2005.PubMed/NCBI View Article : Google Scholar

46 

Graves JA, Wang Y, Sims-Lucas S, Cherok E, Rothermund K, Branca MF, Elster J, Beer-Stolz D, Van Houten B, Vockley J and Prochownik EV: Mitochondrial structure, function and dynamics are temporally controlled by c-Myc. PLoS One. 7(e37699)2012.PubMed/NCBI View Article : Google Scholar

47 

Miller DM, Thomas SD, Islam A, Muench D and Sedoris K: c-Myc and cancer metabolism. Clin Cancer Res. 18:5546–5553. 2012.PubMed/NCBI View Article : Google Scholar

48 

Leites EP and Morais VA: Mitochondrial quality control pathways: PINK1 acts as a gatekeeper. Biochem Biophys Res Commun. 500:45–50. 2018.PubMed/NCBI View Article : Google Scholar

49 

Panigrahi DP, Praharaj PP, Bhol CS, Mahapatra KK, Patra S, Behera BP, Mishra SR and Bhutia SK: The emerging, multifaceted role of mitophagy in cancer and cancer therapeutics. Semin Cancer Biol. 66:45–58. 2020.PubMed/NCBI View Article : Google Scholar

50 

Wang H, Ni HM, Chao X, Ma X, Rodriguez YA, Chavan H, Wang S, Krishnamurthy P, Dobrowsky R, Xu DX, et al: Double deletion of PINK1 and Parkin impairs hepatic mitophagy and exacerbates acetaminophen-induced liver injury in mice. Redox Biol. 22(101148)2019.PubMed/NCBI View Article : Google Scholar

51 

Rak M, Bénit P, Chrétien D, Bouchereau J, Schiff M, El-Khoury R, Tzagoloff A and Rustin P: Mitochondrial cytochrome c oxidase deficiency. Clin Sci (Lond). 130:393–407. 2016.PubMed/NCBI View Article : Google Scholar

52 

Gundamaraju R, Lu W and Manikam R: Revisiting Mitochondria Scored Cancer Progression and Metastasis. Cancers (Basel). 13(432)2021.PubMed/NCBI View Article : Google Scholar

53 

Vieira AF and Schmitt F: An update on breast cancer multigene prognostic tests-emergent clinical biomarkers. Front Med (Lausanne). 5(248)2018.PubMed/NCBI View Article : Google Scholar

54 

Gogvadze V, Orrenius S and Zhivotovsky B: Mitochondria as targets for cancer chemotherapy. Semin Cancer Biol. 19:57–66. 2009.PubMed/NCBI View Article : Google Scholar

55 

Leber B, Geng F, Kale J and Andrews DW: Drugs targeting Bcl-2 family members as an emerging strategy in cancer. Expert Rev Mol Med. 12(e28)2010.PubMed/NCBI View Article : Google Scholar

56 

Indran IR, Tufo G, Pervaiz S and Brenner C: Recent advances in apoptosis, mitochondria and drug resistance in cancer cells. Biochim Biophys Acta. 1807:735–745. 2011.PubMed/NCBI View Article : Google Scholar

57 

Dijk SN, Protasoni M, Elpidorou M, Kroon AM and Taanman JW: Mitochondria as target to inhibit proliferation and induce apoptosis of cancer cells: The effects of doxycycline and gemcitabine. Sci Rep. 10(4363)2020.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

October-2021
Volume 15 Issue 4

Print ISSN: 2049-9450
Online ISSN:2049-9469

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lebok P, Schütt K, Kluth M, Witzel I, Wölber L, Paluchowski P, Terracciano L, Wilke C, Heilenkötter U, Müller V, Müller V, et al: High mitochondrial content is associated with breast cancer aggressiveness. Mol Clin Oncol 15: 203, 2021
APA
Lebok, P., Schütt, K., Kluth, M., Witzel, I., Wölber, L., Paluchowski, P. ... Burandt, E. (2021). High mitochondrial content is associated with breast cancer aggressiveness. Molecular and Clinical Oncology, 15, 203. https://doi.org/10.3892/mco.2021.2365
MLA
Lebok, P., Schütt, K., Kluth, M., Witzel, I., Wölber, L., Paluchowski, P., Terracciano, L., Wilke, C., Heilenkötter, U., Müller, V., Schmalfeldt, B., Simon, R., Sauter, G., Von Leffern, I., Krech, T., Krech, R. H., Jacobsen, F., Burandt, E."High mitochondrial content is associated with breast cancer aggressiveness". Molecular and Clinical Oncology 15.4 (2021): 203.
Chicago
Lebok, P., Schütt, K., Kluth, M., Witzel, I., Wölber, L., Paluchowski, P., Terracciano, L., Wilke, C., Heilenkötter, U., Müller, V., Schmalfeldt, B., Simon, R., Sauter, G., Von Leffern, I., Krech, T., Krech, R. H., Jacobsen, F., Burandt, E."High mitochondrial content is associated with breast cancer aggressiveness". Molecular and Clinical Oncology 15, no. 4 (2021): 203. https://doi.org/10.3892/mco.2021.2365