Academic Editor: Enrique Hernandez
Triple-negative breast cancer (TNBC) lacks expression of the three biomarkers (the estrogen receptor (ER), progesterone receptor (PR), and the human epidermal growth factor receptor 2 (HER2) protein) and are typically higher grade. While the triple-negative clinical phenotype is heterogeneous, the basal-like molecular subtype comprises a large proportion, particularly for breast cancer susceptibility gene 1 (BRCA1)-associated breast cancer. New treatment options are checkpoint inhibitors like inhibition of PD-L1 pathway with pembrolizumab and atezolizumab, parp-inhibition with olaparib or talozoparib and treatment with the an antibody drug conjugate sacituzumab-govitecan.
Triple-negative breast cancer (TNBC) describes breast cancers that lack
expression of the estrogen receptor (ER), progesterone receptor (PR), and human
epidermal growth factor receptor 2 (HER2). TNBC behaves more aggressively than
other types of breast cancer. Although immunotherapy (in combination with
chemotherapy) is available for advanced TNBC that expresses programmed cell death
ligand 1 (PD-L1), there are no approved targeted treatments in TNBC comparing
with other breast cancer subtypes (i.e., ER-positive, HER2-positive subtypes). For
purposes of this review, we consider “triple-negative” to mean cancers that
have
A more extensive discussion on surgical management, neoadjuvant chemotherapy, adjuvant chemotherapy of non-metastatic breast cancer, and the treatment of metastatic breast cancer is covered separately.
EPIDEMIOLOGY — TNBC accounts for approximately 15 percent of breast cancers diagnosed worldwide — almost 200,000 cases each year [4]. Compared with hormone receptor-positive breast cancer, TNBC is more commonly diagnosed in women younger than 40 years. In one study, there was a twofold higher attributable risk of TNBC in women under 40 years compared with women over 50 years (odds ratio (OR) 2.13, 95% confidence interval (CI) 1.34–3.39) [5]. In addition, TNBC appears to be relatively more common among black women compared with white women (OR 2.41, 95% CI 1.81–3.21) [5]. It is important to mention that different molecular subtypes of TNBC like basal-like 1 (BL1), basal-like 2 (BL2), immunomodulatory (IM), mesenchymal (M), mesenchymal stem–like (MSL) and luminal androgen receptor (LAR) subtype are described with different prognostic impact and oncological behaviour [6].
Risk factors associated with the diagnosis of TNBC include:
BRCA testing—In light of the association of particular breast cancer susceptibility gene 1 (BRCA1) mutations with TNBC, we recommend that women diagnosed at 60 years or younger with a localized TNBC, or those of any age with metastatic TNBC, undergo BRCA mutation testing regardless of family history (See “Genetic testing and management of individuals at risk of hereditary breast and ovarian cancer syndromes”).
For those with metastatic disease, results of BRCA testing have therapeutic implications (See ‘Metastatic disease’ below).
The neoadjuvant or adjuvant chemotherapy options for patients with TNBC are similar to the approaches used in other breast cancer phenotypes. The principles for the surgical management of and radiation therapy options for breast cancer are also applied in a similar way across breast cancer subtypes (See “Breast-conserving therapy” and “The role of local therapies in metastatic breast cancer” and “Adjuvant radiation therapy for women with newly diagnosed, non-metastatic breast cancer” and “Radiation therapy techniques for newly diagnosed, non-metastatic breast cancer”).
Chemotherapy—Chemotherapy is recommended for women with TNBC
Neoadjuvant versus adjuvant administration — Neoadjuvant chemotherapy (NACT) is the preferable approach in patients with locally advanced breast cancer or for those who are not candidates for or unlikely to have a good cosmetic outcome with breast conservation. For patients receiving NACT, pathologic complete response is associated with improvement in disease-free survival (DFS) [30, 31, 32]. Additionally, patients with smaller (e.g., T1c) TNBCs may be offered neoadjuvant therapy, particularly if they might be candidates for additional treatments in the adjuvant setting if residual disease is identified. The approach to neoadjuvant therapy for patients with breast cancer, including further discussion of appropriate candidates, with special considerations for those with TNBC, is found elsewhere (See “General principles of neoadjuvant management of breast cancer” and “General principles of neoadjuvant management of breast cancer”, section on ‘Patient selection’ and “Choice of neoadjuvant chemotherapy for HER2-negative breast cancer”, section on ‘Special considerations for triple-negative disease’).
The role for additional chemotherapy in the adjuvant setting for women with residual cancer after neoadjuvant chemotherapy is discussed elsewhere.
Benefits—In general, there is a larger absolute benefit to adjuvant chemotherapy among patients with TNBC compared with those with hormone-positive disease [33].
An analysis of three randomized trials with a total of 6644 women with node-positive breast cancer comparing patients those with ER-positive breast cancer with those with ER-negative breast cancer showed the following significant outcomes at five years following adjuvant chemotherapy [33]:
These data emphasize the importance of neo/adjuvant chemotherapy for women with TNBC, who (unlike those with ER-positive or HER2-positive breast cancer) are not eligible for targeted therapies.
For example, in a randomized trial of nearly 650 patients with operable TNBC, those assigned to six cycles of adjuvant paclitaxel and carboplatin (administered on days 1, 8, and 15 every 28 days) had a longer DFS relative to those assigned to an anthracycline and taxane based regimen (five-year DFS 87 versus 80%), with similar OS [38].
However, for patients with stage I disease, adjuvant rather than neoadjuvant treatment is appropriate, using standard regimens such as AC-T or TC. In general, for patients who have not received neoadjuvant chemotherapy, adding antimetabolite agents such as capecitabine or gemcitabine to adjuvant chemotherapy has not improved OS outcomes in TNBC [40, 41], and it is not our approach. A Chinese trial demonstrated improvement in DFS, but not OS, with capecitabine following standard adjuvant regimens [42]. Among 434 women with early-stage TNBC who received standard adjuvant treatment (94% of whom had not received neoadjuvant therapy), low-dose capecitabine maintenance therapy for one year improved five-year DFS compared with observation only (83 versus 73%; hazard ratio (HR) 0.64, 95% Confidence interval (CI) 0.42–0.95). The five-year OS was similar between the groups (86 versus 81%, with and without capecitabine, respectively; HR 0.75, 95% CI 0.47–1.19). The trial had important limitations; notably, there was an imbalance in randomization, with a higher proportion of older women assigned to placebo, which could have favored the capecitabine group.
Another phase III trial of 876 women with early-stage TNBC demonstrated that the subsequent treatment with capecitabine after standard adjuvant chemotherapy versus placebo resulted in numerically, but not statistically, improved five-year DFS and OS (DFS, 80% versus 77%, HR 0.79, 95% CI 0.61–1.03; OS, 86.2 versus 85.9%, HR 0.92, 95% CI 0.66–1.28) [40]. Similarly, trials looking at adjuvant gemcitabine have proven negative.
Given the sum of data, we opt for standard anthracycline- and/or taxane-based chemotherapy regimens as adjuvant therapy in patients with TNBC who have not received neoadjuvant treatment. As discussed, in practice, only lower-risk patients (i.e., stage I TNBC) are treated with adjuvant rather than neoadjuvant chemotherapy, as most patients with higher-risk disease receive neoadjuvant therapy.
Treatment of tumors
In a retrospective review of almost 4400 patients with small, node-negative TNBCs (6.5 percent with pT1a, 21 percent with pT1b, and 72 percent with pT1c tumors), 53% of patients received adjuvant chemotherapy [45]. These patients had more unfavorable baseline characteristics including younger age, larger tumors, and higher tumor grade. A multivariate analysis showed, that adjuvant chemotherapy improved breast cancer-specific survival in the overall group (adjusted HR 0.65, 95% CI 0.48–0.89), but not for the subset of patients with pT1a tumors (adjusted HR 4.28, 95% CI 1.12–16.44). Although limitations of this study include its retrospective nature and that the number of patients with pT1a tumors was small (n = 18), the results suggest that the risks of chemotherapy may outweigh benefits in patients with these small tumors.
The natural history of small triple-negative tumors was demonstrated in a study of 143 patients with triple-negative tumors up to 1 cm in size and not treated with adjuvant chemotherapy [46]. Patients with triple-negative tumors had a 75 to 89 percent relapse-free survival and over 95 percent distant relapse-free survival at five years. Another study including 363 T1a-bN0 triple-negative tumors from the National Comprehensive Cancer Network (NCCN) database suggested a 90 to 93 percent distant relapse-free survival without chemotherapy [47]. Given the lack of prospective data on women who present with small tumors, the decision to administer adjuvant chemotherapy must be individualized based on patient and provider preferences.
Prognosis—The peak of the risk of distant recurrence and death is approximately three years after diagnosis declining rapidly thereafter [31]. TNBC is characterized by higher relapse rates during this period of time compared with ER-positive breast cancers, although the latter tend to continue to recur for decades later while TNBCs tend not to do so. Therefore, overall in the long run the absolute risk of recurrence for the two subtypes approach one another. Furthermore, however, TNBC may be more likely to recur in locoregional areas as well as in visceral organs, such as liver, lung, and brain involvement at first recurrence [48, 49, 50, 51]. By contrast, TNBC is less likely than ER-positive breast cancer to recur initially in bone [51]. In one study involving 116 patients with triple-negative metastatic breast cancer, brain metastases were the initial site of metastatic disease or occurred during their metastatic course in 14 and 46 percent, respectively [49]. The median survival following a diagnosis of central nervous system metastases is less than six months [52, 53].
Patients with TNBC have a poorer short-term (first five to seven years) prognosis compared with patients with other breast cancer subtypes [15, 26, 51, 54]. In a 2012 study of 12,902 women who presented to NCCN centers, compared with women with hormone receptor-positive, HER2-negative breast cancer, women with TNBC experienced, at a median follow-up of three years [51]:
The risk of late recurrence is low for women with TNBC. In a single-center retrospective series of 783 women with stage I, II, or III TNBC who were alive and without recurrence at five years after treatment for the original diagnosis, the yearly recurrence-free interval at 10 and 15 years was 97 and 95 percent, respectively, and the relapse-free survival rates were 91 and 83 percent, respectively [55]. In a prospective cohort study in which patients with stage I to III breast cancer diagnosed between 1986 and 1992 were matched with patients diagnosed between 2004 and 2008, the hazard rate of relapse for those with triple-negative disease had dropped to essentially zero after year 6 among patients treated in the later cohort [56].
Post-treatment surveillance — There are no specific post-treatment surveillance guidelines for patients with TNBC. Patients with breast cancer should undergo a similar surveillance routine according to American Society of Clinical Oncology guidelines following breast cancer treatment, regardless of breast cancer subtype. This should include history and complete physical exam every three months for the first three years, then every 6 to 12 months for surveillance. A further discussion on post-treatment surveillance is covered separately (See “Approach to the patient following treatment for breast cancer”, section on ‘Guidelines for post-treatment follow-up’).
Many of the general principles applicable to advanced breast cancer of other phenotypes apply to that of TNBC. The cornerstone of systemic treatment for TNBC has been chemotherapy because endocrine and HER2-directed therapies are ineffective. However, several trials have suggested a role for targeted therapies in TNBC including inhibitors of poly(ADP-ribose) polymerase (PARP) and immune checkpoints (See “Systemic treatment for metastatic breast cancer: General principles” and “Systemic treatment of metastatic breast cancer in women: Chemotherapy”).
Repeat biopsy—In patients with metastatic breast cancer, a confirmatory biopsy of a suspected lesion should be obtained when possible, with the following assessments:
Because the US Food and Drug Administration (FDA) has approved each test as a “companion diagnostic” with a specific immune checkpoint inhibitor rather than approval as a class, either of the companion diagnostics is acceptable.
However, if needed, these assessments can instead be performed in a subsequent biopsy after progression, given that they will not dictate choice of initial therapy and that these abnormalities are relatively rare in breast cancer.
In addition to these assays performed on tissue biopsy, all patients with TNBC should undergo genetics evaluation to determine if they are BRCA carriers, given the therapeutic implications in advanced disease (See ‘No germline BRCA mutation’ below and ‘Germline BRCA mutation’ below).
Initial treatment for rapidly progressive visceral disease—Combination chemotherapy may be appropriate for those with extensive or rapidly progressive visceral disease, in whom the higher chance of response is thought to outweigh the higher risks of toxicity, due to concerns about impending organ dysfunction. However, both clinicians and patients should know there are no prospective data that show combination chemotherapy improves overall survival (OS) compared with single-agent sequential cytotoxic chemotherapy. Further details are discussed elsewhere (See “Systemic treatment of metastatic breast cancer in women: Chemotherapy”, section on ‘Combination chemotherapy’).
Initial treatment in the absence of rapidly progressive visceral disease — As discussed above, patients with metastatic TNBC should have germline testing for BRCA, as well as tumor assessment for PD-L1 (See ‘Genetics evaluation’ above and ‘Repeat biopsy’ above).
The initial treatment approach depends on the outcomes of these assessments.
PD-L1-negative tumors—Our approach to most patients with advanced, sporadic, triple-negative metastatic breast cancer that does not express programmed cell death ligand 1 (PD-L1) is to use single-agent chemotherapy. However, combination chemotherapy strategies may be appropriate in some such patients with rapidly progressive visceral disease (See “Systemic treatment of metastatic breast cancer in women: Chemotherapy”, section on ‘Single-agent chemotherapy’ and “Systemic treatment of metastatic breast cancer in women: Chemotherapy”, section on ‘Combination chemotherapy’).
Either platinum- or non-platinum-based regimens are appropriate, with a choice driven by toxicity profiles. A meta-analysis with 10 randomized trials comparing platinum-containing chemotherapy with regimens not containing platinum in 958 cases of metastatic TNBC demonstrated, that the death rate in the platinum group was 46% versus 51% in the non-platinum group (hazard ratio (HR) 0.85, 95% CI 0.73–1.00) at one year [62]. However, the platinum recipients complained more grade 3 and 4 toxicities, including nausea/vomiting (relative risk (RR) 4.8) and anemia (RR 3.8).
Outcomes of platinum and non-platinum regimens in breast cancer susceptibility gene (BRCA)-associated TNBCs are discussed below (See ‘Chemotherapy-naive patients, or those with progression on PARP inhibitors’ below).
For those with tumor expression of programmed cell death ligand 1 (PD-L1), we recommend the addition of an immune checkpoint inhibitor to chemotherapy, rather than chemotherapy alone.
The checkpoint inhibitor atezolizumab is European Medicines Agency (EMA) approved for use with nabpaclitaxel
for those with advanced TNBC with PD-L1-stained, tumor-infiltrating immune cells
of any intensity covering
Although these therapies are approved irrespective of treatment line, the supporting data were based on patient experiences receiving first-line treatment for metastatic disease. The benefits as later-line treatment for metastatic disease are not known. Recognizing this limitation, patients with prior taxane treatment (either in the (neo)adjuvant or metastatic setting) are still candidates for the atezolizumab/nabpaclitaxel combination.
Overall, at a median follow-up of 13 months, there was only a modest but statistically significant difference in progression-free survival (PFS) in favor of incorporating atezolizumab. PFS for those receiving atezolizumab versus those who did not was 7.2 versus 5.5 months (HR for progression or death 0.80, 95% CI 0.69–0.92), with a nonsignificant trend towards improved OS (21.3 versus 17.6 months; HR for death 0.84, 95% CI 0.69–1.02).
However, a prospectively planned subset analysis of outcomes according to PD-L1-expression showed, that atezolizumab improved both PFS (7.5 versus 5 months; HR 0.62, 95% CI 0.49–0.78), and OS (25 versus 15.5 months; HR 0.62, 95% CI 0.45–0.86). Final OS analysis at 20 months’ follow-up demonstrated continued improved survival in the PD-L1-positive subset with the addition of atezolizumab to nabpaclitaxel (median OS (95% CI): 17.9 months (13.6–20.3) versus 25.4 months (19.6–30.7); stratified HR (95% CI): 0.67 (0.53, 0.86)) and similar adverse events, with 23 percent experiencing thyroid disease and approximately 10 percent with other immune-related adverse events. But it has to be mentioned OS analysis was not formally tested for statistical significance [65].
Another trial, IMpassion 131, examined atezolizumab combined with paclitaxel in first-line metastatic TNBC, with a focus on PD-L1-positive tumors defined similarly to IMpassion 130. However, unlike IMpassion 130, no significant improvement in PFS in the PD-L1-positive subset was observed, at just under nine months’ follow-up (5.7 versus 6 months) [66]. The explanation for the discordance in results is unclear; however, given that the main difference between IMpassion 130 and 131 was the chemotherapy backbone, the preferred combination with atezolizumab remains nabpaclitaxel.
In addition to the chemotherapy combination trials noted above, early clinical
experience with immunotherapy (pembrolizumab (anti PD-1 anibody); avelumab and
atezolozumab (anti PD-L1-antibody)) in the setting of TNBC shows response rates
Patients with previous exposure to chemotherapy — Inhibitors of PARP may be particularly useful in breast cancer susceptibility gene (BRCA)-mutated breast cancers, of which the majority are triple negative. For most patients with TNBC with germline BRCA mutations who have previously been treated with chemotherapy in the neoadjuvant, adjuvant, or metastatic disease setting, we suggest an oral inhibitor of PARP rather than chemotherapy, since the data suggest improved efficacy and fewer side effects. However, chemotherapy is appropriate if and when a patient suffers progressive disease on a PARP inhibitor; or for those who are chemotherapy naive, having never received chemotherapy either in the early-stage or metastatic setting; or, as discussed above, for those with rapidly progressive visceral disease (See ‘Initial treatment for rapidly progressive visceral disease’ above).
Additionally, the combination of immunotherapy and chemotherapy is an acceptable alternative to a PARP inhibitor for those with PD-L1-positive disease (See ‘Chemotherapy-naive patients, or those with progression on PARP inhibitors’ below and ‘PD-L1-positive tumors’ above).
In the OlympiAD trial (subset of 121 BRCA mutation carriers with metastatic triple-negative disease having been treated with an anthracycline and a taxane in either the adjuvant or metastatic setting) patients receiving olaparib experienced an improved PFS (HR 0.43, 95% CI 0.29–0.63) [72]. Compared with hormone receptor-positive, HER2-negative patients the triple negative patients had a greater beneifit from olaparib treatment. In the TNBC gBRCA mutated subgroup of the EMBRACA trial, talazoparib also improved PFS (HR 0.60, 95% CI 0.41–0.87). Further details of these studies are discussed elsewhere. It should be noted that the comparator single-agent chemotherapy options did not include either taxanes or platinums in these studies, so the trial realistically only compared PARP inhibitors against second-line therapies. It is unknown how PARP inhibitors would compare with first-line drugs (See “Systemic treatment for metastatic breast cancer: General principles”, section on ‘PARP inhibition for BRCA carriers’).
There are several other PARP inhibitors in clinical development [73, 74, 75, 76, 77, 78, 79]. For example, veliparib (ABT-888) was evaluated (single arm phase II trial) in combination with the alkylating agent temozolomide in a group of 41 women with advanced TNBC (of whom 8 had a BRCA germline mutation) [79]. While the overall response and clinical benefit rates were 7 and 17% across the entire study population, a clear improvement was noticed in patients with BRCA mutations with an overall response and clinical benefit rates of 37.5 and 62.5%, respectively. The results of the ISPY trial that evaluated the combination of veliparib plus carboplatin when combined with standard chemotherapy as part of a neoadjuvant treatment program in women with TNBC are discussed elsewhere (See “Choice of neoadjuvant chemotherapy for HER2-negative breast cancer”, section on ‘Special considerations for triple-negative disease’).
There is mechanistic rationale for use of PARP inhibition as anticancer therapy. PARP is involved in the molecular events leading to cell recovery from DNA damage. The inhibition of PARP1 leads to an accumulation of double-strand DNA breaks. Normally, these breaks are repaired by the BRCA pathway-dependent homologous recombination mechanism [80]. There is the hypothesis that the combination treatment of PARP inhibition with DNA-damaging chemotherapeutics would affect tumors lacking BRCA function [73, 81, 82, 83].
Chemotherapy-naive patients, or those with progression on PARP inhibitors;
Although we typically start with a poly (ADP-ribose) polymerase (PARP) inhibitor for metastatic disease in those with germline BRCA mutations who have had chemotherapy in the (neo)adjuvant setting, chemotherapy is the preferred option for those who have never been exposed to chemotherapy (in the early or metastatic settings); or for those who have experienced progression on a PARP inhibitor; or for those with rapidly progressive visceral disease, as discussed above.
When administering chemotherapy, our approach is as follows:
The TNT randomized trial directly compared carboplatin and docetaxel in the first-line treatment setting for women with metastatic TNBC. Overall response rates were similar in the overall group, but among the 43 women with a known BRCA1/2 mutation, carboplatin resulted in a higher response rate (68 versus 33%; absolute difference 35 percent, 95% CI 6.3–63.1%) and PFS (6.8 versus 4.4 months; absolute difference 2.6 months, 95% CI 0.11–5.12 months) [85]. However, the trial had a crossover design, and no statistically significant OS difference was seen (12.8 months, 95% CI 10.6–15.3; and 12 months, 95% CI 10.2–13) for those allocated carboplatin or docetaxel, respectively, suggesting that either agent may be administered first, without compromising outcomes.
Grade
Trop-2 is expressed in the majority of TNBCs. Sacituzumab govitecan is an antibody-drug conjugate that targets Trop-2 for the selective delivery of SN-38, the active metabolite of irinotecan. It is approved by the Food and Drug Administration (FDA) for the treatment of adult patients with metastatic TNBC who have received at least two prior therapies for metastatic disease [86]. Severe neutropenia and diarrhea may occur with this agent, including cases of neutropenic colitis. Management of enterotoxicity of this agent is discussed elsewhere.
In a single-arm trial of 108 patients with previously treated metastatic TNBC
(median of three previous treatments), the objective response rate to sacituzumab
govitecan was 34 percent, with a median PFS of 5.5 months, duration of response
of 9.1 months, and OS of 13 months [87]. Grade
Pembrolizumab for tumors with high TMB or MSI-H/dMMR tumors—The immune
checkpoint inhibitor pembrolizumab is approved by the FDA for the treatment of
unresectable or metastatic, microsatellite instability-high (MSI-H) or mismatch
repair-deficient (dMMR) solid tumors, as well as tumors with high tumor
mutational burden (TMB;
We acknowledge, in discussion with patients, that the trials supporting the approval of pembrolizumab for these indications did not include breast cancer patients, but efficacy was demonstrated in other cancer types, including cervical, endometrial, and ovarian cancer.
Several promising treatment options are under active clinical investigation but should not be used at this time outside of a clinical trial. These are discussed below.
Triple-negative breast cancer (TNBC) lacks expression of the three most commonly evaluated biomarkers (the estrogen receptor (ER), progesterone receptor (PR), and the human epidermal growth factor receptor 2 (HER2) protein) and are both typically higher grade and are more likely to be diagnosed clinically rather than mammographically than ER-positive cancers. While the triple-negative clinical phenotype is heterogeneous, the basal-like molecular subtype comprises a large proportion, particularly for breast cancer susceptibility gene 1 (BRCA1)-associated breast cancer.
In the non-metastatic disease the principles that apply to the surgical
treatment and use of radiation therapy in breast cancer, and the systemic
treatment approach in both the neoadjuvant and adjuvant settings, are similar in
TNBC and other HER2-negative subtypes. For patients with TNBC and either a tumor
size
In the metastatic setting, combination chemotherapy may be appropriate for those
with extensive or rapidly progressive visceral disease, in whom the higher chance
of response is thought to outweigh the higher risks of toxicity. However, there
are no prospective data that show combination chemotherapy improves overall
survival (OS) compared with single-agent sequential cytotoxic chemotherapy. In
the metastatic TNBC setting, for those who are not in visceral crisis, therapy
depends on prior treatment history, programmed cell death ligand 1 (PD-L1)
expression, and germline BRCA mutation status. For PD-L1-positive TNBC in
BRCA-wildtype patients, as well as in chemotherapy-naive BRCA carriers, the
combination of an immune checkpoint inhibitor and chemotherapy as initial
treatment for metastatic disease is recommended rather than single-agent
chemotherapy (Grade 1B). The checkpoint inhibitor atezolizumab is EMA approved
for use with nabpaclitaxel in advanced TNBC with PD-L1
MF, SK, MW and DS designed the manuscript idea. MF, SK, MW and DS analyzed the literature data. DF, CR, GR and CK reviewed the manuscript. DF, CR, GR and CK edited the manuscript. All authors contributed to editorial changes in the manuscript. All authors read and approved the final manuscript.
Not applicable.
We would like to express our gratitude to all those who helped us during the writing of this manuscript. Thanks to all the peer reviewers for their opinions and suggestions.
This research received no external funding.
MF is serving as one of the Editorial Board members of this journal and the guest editor for the special issue titled “Breast Cancer”. We declare that MF had no involvement in the peer review of this article and has no access to information regarding its peer review. Full responsibility for the editorial process for this article was delegated to Enrique Hernandez. The other authors declare no conflict of interest.