HER2-Positive Breast Cancer: History
Please note this is an old version of this entry, which may differ significantly from the current revision.

HER2 is a member of the epidermal growth factor (EGF) receptor family comprising EGFR/ErbB1, HER2/ErbB2, HER3/ErbB3, and HER4/ErbB4. EGF receptor family proteins encode for transmembrane receptors consisting of the extracellular ligand binding, lipophilic transmembrane, and cytoplasmic tyrosine kinase domains. Ligand binding to EGF receptors activates their hetero- or homodimerization whereby one EGF receptor will phosphorylate the other on a tyrosine, allowing cytoplasmic signaling complexes to bind to the EGFR dimer.

  • mTOR
  • glycolysis
  • HER2-positive breast cancer
  • 2-deoxyglucose
  • trastuzumab
  • Herceptin
  • LKB1
  • tumor recurrence
  • rapalog
  • PI3K-Akt-mTOR signaling pathway
  • clinical diagnostics

1. Introduction

In women, the most diagnosed cancer and a leading cause of cancer-related deaths worldwide is breast cancer [1,2]. The type of breast cancer is characterized by overexpression or absence of hormonal receptors: estrogen receptor-positive (ER+), progesterone-positive (PR+), human epidermal growth factor receptor 2-positive (HER2-positive), or the absence of ER/PR/HER2 (triple-negative). HER2-positive breast cancer constitutes 15–30% of all breast tumors [7,8,9]. Of these subtypes, HER2-E constitutes about 50–60% of all HER2-positive breast cancers [11,12,13]. HER2-positive breast tumors progress faster and more aggressively than most other breast tumors. Anti-HER2 therapies often lead to the development of chemoresistance (reviewed in [14]) and an elevated risk of recurrence that increases mortality rates [15] (reviewed in [16]). HER2-positive breast cancer is associated with hyperactivation of the mTOR pathway and a metabolic shift from aerobic respiration to glycolysis. As the mechanistic target of rapamycin (mTOR) pathway [17] and glycolysis [18] also contribute to supporting tumor recurrence and chemoresistance, these signaling pathways have become appealing targets for HER2-positive breast cancer therapy.

2. Neu/HER2 in Breast Cancer

HER2 is a member of the epidermal growth factor (EGF) receptor family comprising EGFR/ErbB1, HER2/ErbB2, HER3/ErbB3, and HER4/ErbB4. EGF receptor family proteins encode for transmembrane receptors consisting of the extracellular ligand-binding, lipophilic transmembrane, and cytoplasmic tyrosine kinase domains. However, the proto-oncogene Neu/HER2 (HER2) has no known natural ligands [21]; thus, to elicit downstream signaling, HER2 preferentially dimerizes with EGF receptors that subsequently results in a more potent signaling response. Due to its role in activating signaling pathways that stimulate cell proliferation and survival, HER2 overexpression in breast tumor cells promotes tumor growth and increases the percentage of cells in S phase, aneuploidy, and migration of cancer cells into the lymph nodes [22] (reviewed in [23]).

Rather, trastuzumab binding to HER2 induces antibody-dependent cellular cytotoxicity (ADCC) in which HER2-bound trastuzumab directs natural killer (NK) cells to destroy tumor cells [30]. Furthermore, HER2-specific monoclonal antibodies directed towards several epitopes were effective at reducing tumor growth in vivo [31] and enhanced the recruitment of Cbl to phosphorylated HER2 (Y1112), followed by its proteasomal degradation [31]. While the preliminary phase II trials observed minimal adverse cardiac effects [32,33], a phase III trial using patients, of whom many had previously received anthracycline therapy, had a high rate of cardiac dysfunction [37]. Evidence later indicated that the adverse cardiac effects were fairly low for trastuzumab monotherapy and prior anthracycline treatment (3–7%) and anthracycline plus cyclophosphamide (8%), but trastuzumab and anthracycline plus cyclophosphamide treatment showed a much higher incidence (27%) [38].

[61] evaluated the therapeutic ability of lapatinib using a panel of 31 breast cancer cell lines that also included trastuzumab-resistant HER2-positive breast cancer cells. Lapatinib treatment of HER2-positive breast cancer cell lines was shown to inhibit HER2 and EGFR activity as well as downstream phosphorylation of Akt and ERK. Xenograft mouse models implanted with HER2-positive breast cancer cells showed a significant reduction in tumor volume in the lapatinib treatment group compared to the control group. Despite the clinical benefit of lapatinib, resistance develops through re-activation of mTOR signaling, and up-regulation of nuclear receptor ERRα, a key regulator of cell metabolism that is normally degraded in response to lapatinib treatment [62].

[63] examined the effects of neratinib using human HER2-positive breast cancer cell lines, where it was found neratinib treatment down-regulated MAPK, Akt, and RB phosphorylation, down-regulated cyclin D1 expression, and up-regulated the cell cycle inhibitor p27 in a dose-dependent manner. Furthermore, preclinical experiments showed that neratinib treatment reduced tumor growth in xenograft mouse models implanted with HER2-positive breast cancer cells compared to the controls [63,64]. Phase II trials observed high response rates and median PFS in neratinib treatment of HER2-positive breast cancer, but these were lower in patients with prior trastuzumab treatment. In the phase III NALA trial, patients previously treated for metastatic HER2-positive breast cancer responded more favorably to neratinib plus capecitabine, particularly HER2-positive breast cancer patients that were hormone receptor-negative (HR−)

They found that monotherapy of pertuzumab reduced the tumor volume to a similar extent as the trastuzumab treatment, but tumor growth was significantly quenched with dual therapy of both monoclonal antibodies. Phase II studies found pertuzumab/trastuzumab combined therapy was successful in treating HER2-positive breast cancer, with more than 10% of patients experiencing adverse effects, but the therapy was otherwise well-tolerated [49,50,51]. In the APHINITY clinical trials [52,70], patients with early HER2-positive breast cancer were treated with pertuzumab or placebo in combination with trastuzumab and chemotherapy. II studies, the addition of pertuzumab to a trastuzumab and chemotherapy treatment regimen did not result in any significant increase in adverse effects other than elevated diarrhea scores [70].

Trastuzumab emtansine (trastuzumab-DM1) is an antibody-drug conjugate where trastuzumab is stably bound to DM1, a derivative of anti-tumor drug maytansine. In phase II clinical trials, trastuzumab-DM1 monotherapy showed similar success compared to other HER2 therapies in HER2-positive breast cancer and was well-tolerated with only ~20% of patients experiencing adverse effects [53]. In phase II clinical trials, DS-8201 showed an ORR of 60.2% in patients with HER2-positive metastatic breast cancer [55]. Although gastrointestinal and hematologic toxicity was a common occurrence in patients during treatment, no cardiotoxicity was observed, but an increased risk of interstitial lung disease was associated with treatment.

Like lapatinib, tucatinib binds to the ATP pocket of HER2 and acts as a competitive, reversible tyrosine kinase inhibitor, but tucatinib is selective for HER2 only [73]. [73] also determined that tucatinib effectively inhibited HER2 phosphorylation in vitro using the HER2-positive breast cancer cell line BT-474 and observed a minimal inhibition of EGFR phosphorylation in the EGFR-overexpressing skin cancer cell line A431. In xenograft mouse models of HER2-positive breast cancer implanted with BT-474 cells, tucatinib treatment resulted in a delay in tumor growth comparable to trastuzumab monotherapy, and these effects were enhanced in the combination therapy of tucatinib plus trastuzumab [73]. In HER2CLIMB, a phase II clinical trial, tucatinib–trastuzumab–capecitabine combination therapy for HER2-positive breast cancer showed a higher ORR and median PFS than the placebo–trastuzumab–capecitabine group; however, this was associated with a high incidence of adverse effects [56].

Pyrotinib (or SHR1258) is an irreversible inhibitor of EGFR/HER1, HER2, and HER4 shown to suppress tumor growth in HER2-positive breast cancer xenograft mouse models, which was associated with a favorable safety profile [74]. In phase I and II clinical trials, Ma et al. [57,58] reported that the combination therapy of pyrotinib and capecitabine resulted in significantly improved ORR and PFS as compared to lapatinib and capecitabine therapy. Pyrotinib treatment was also well-tolerated, with a low percentage of patients experiencing grade 3 adverse events.

Although success using trastuzumab to treat HER2-positive breast cancer has been achieved, the expensiveness of this drug limits accessibility to patients. Hence, biosimilar drugs ameliorate this issue as they are nearly indistinguishable from the original drug and are less expensive. when compared to trastuzumab [76]. [59] reported that HXL02 showed similar efficacy and adverse effects in patients with HER2-positive recurrent or metastatic breast cancer compared to patients receiving trastuzumab.

3. The mTOR Pathway in HER2-Positive Breast Cancer

Enhanced activity of mTOR is associated with HER2-overexpressing breast cancers [77,78]. mTORC1 is composed of five components: mTOR, regulatory-associated protein of mTOR (raptor), mammalian lethal with Sec13 protein 8 (mLST8 or GβL), Like mTORC1, mTORC2 also contains mTOR, mLST8, and deptor, but has the rapamycin-insensitive companion of mTOR (rictor), mammalian stress-activated protein kinase interacting protein (mSIN1), and protein observed with Rictor-1 (Protor-1) subunits [79]. Rictor is unrelated to raptor but facilitates the phosphorylation of various substrates of mTORC2 [81].

AMPK, also a serine-threonine kinase, is a metabolic sensor and negative regulator of the mTOR signaling pathways. AMPK activation is induced by energy-stressed conditions in which intracellular AMP levels are elevated. AMP activates AMPK by binding to the γ subunit and subsequently targeting it for phosphorylation by LKB1 on T172 [86]. In addition, AMPK can directly inhibit mTORC1 by phosphorylation of raptor on S722 and S792 [89].

The PH domain mediates the protein-protein and protein-lipid interactions of Akt. The EXT region contains S473 phosphorylation for full Akt activation (reviewed in [91]). Akt enables mTOR activity by inactivating TSC1/2 through the phosphorylation of TSC2 on several residues, hence enabling Rheb-GTPase to activate mTORC1 [92]. Akt can also directly activate mTORC1 by phosphorylation of mTOR on S2448 [93].

PKA consists of two catalytic subunits and two regulatory subunits and can be activated under glucose-deprived conditions where an accumulation of cellular levels of cAMP occurs. Once activated, PKA promotes the phosphorylation and activation of AMPK in an LKB1-dependent manner [97], which then leads to the inhibition of mTORC1 by phosphorylation of raptor on S791 [98]. [97] found that the knockout of LKB1 significantly reduced AMPK phosphorylation in response to PKA activation; however, the reduction in LKB1 did not completely ablate this effect, possibly due to residual LKB1. They also speculated that AMPK phosphorylation is mediated by another kinase.

mTORC1 activates translational regulators p70 ribosomal S6 kinase 1 (S6K1) and eukaryotic initiation factor 4E (eIF4E)-binding protein 1 (4E-BP1) This prevents incorporation of eIF4E into the eIF4F complex, consequently halting 5’-cap-dependent translation [103]. mTORC1 phosphorylates 4E-BP1 on T37 and T46, which primes 4E-BP1 for subsequent phosphorylation that dissociates it from eIF4E, thereby enabling 5′-cap-dependent translation [104].

Inhibition of mTORC1 activity down-regulated cyclin D1 mRNA and protein levels in MCF7 breast cancer cells, but this effect was rescued when eIF4E activity was enhanced by the knockdown of 4E-BP1. Enhanced activity of eIF4E also increased cyclin D1 translation as knockdown of 4E-BP1 resulted in an increased association of polysomes to the cyclin D1 Furthermore, overexpression of 4E-BP1, in the absence of active mTORC1, led to decreased cyclin D1 levels. A, D1, and E1 were all down-regulated in response to dual inhibition of mTORC1 and mTORC2, knockdown of raptor, and dual knockdown of raptor and rictor.

Although Akt is a positive regulator of mTORC1 activation, mTORC1 negatively regulates Akt activation by modulating its activation by insulin receptor substrate-1 (IRS-1). mTORC1-activated S6K1 inactivates IRS-1 by phosphorylation on S422 [109], leading to its proteasomal degradation [110]. The mTORC1-dependent activation of S6K1 negatively regulates the ERK/MAPK pathway. Inhibition of mTORC1 up-regulated phosphorylated ERK (T202/Y204) levels, which was ablated by the overexpression of constitutively active S6K1 [111].

mTORC1 is involved in glucose uptake and glycolysis by up-regulating the activation of transcription factors such as HIF1α. Knockdown of raptor or inhibition of mTORC1 activity both down-regulated transcript levels of glycolytic enzymesGlut1, Pfkp, andPdk1, which was consistent with the knockdown of HIF1α. Increased glucose uptake due to mTOR hyperactivation was observed inTsc2−/−MEFs, and this effect was blocked by knockdown of HIF1 and inhibition of mTORC1 activity, thus indicating that mTORC1 mediated the increase in glucose uptake through HIF1α. Furthermore, inhibition of mTORC1 also reduced glucose uptake and lactate production in four different AML cells with hyperactive mTOR signaling.

Inhibition of mTORC1 or raptor knockdown both down-regulated transcription of genes involved in oxidative phosphorylation and genes encoding mitochondrial ribosomal proteins and reduced mitochondrial respiration, intracellular ATP levels, and mitochondrial DNA content [117]. [118] observed that the inhibition of mTORC1 resulted in the down-regulation of several genes involved in mitochondrial function and reduced mitochondrial respiration. They found that inhibition of mTORC1 modulated the mRNA levels of several genes comprising the components of complex V of the oxidative phosphorylation pathway, TFAM (a regulator of mitochondrial DNA replication and transcription), numerous mitochondrial ribosomal proteins (MRPLs), and NADH dehydrogenase 1 alpha subcomplex assembly factors 2 and 4 (NDUFAF2 and 4). The knockdown of mTORC1 subunit raptor resulted in a reduction in ATP synthase subunit ATP5O and TFAM expression, mitochondrial respiration, TCA intermediates pyruvate and lactate, and intracellular ATP levels and mitochondrial DNA content as compared to the control.

mTORC1 is a positive regulator of cell growth by repressing autophagy through inhibition of unc-51-like kinase 1 (ULK1) [120], a kinase that initiates autophagy by promoting autophagosome formation. In glucose-starved conditions, ULK1 interacts with AMPK and is subsequently activated by phosphorylation on S317 and S777. In nutrient-sufficient conditions, mTORC1 phosphorylates ULK1 on S757, which prevents autophagosome formation by disrupting the interaction of ULK1 with AMPK [121].

The activation of mTORC1 maintains a feedback loop that inhibits mTORC2 activity. The phosphorylation of rictor did not affect mTORC2 assembly, kinase activity, or cellular localization; however, mutation of T1153 resulted in increased mTORC2 activity [122]. In contrast, the phosphorylation of mSin1 causes the dissociation of mSin1 from mTORC2, thus preventing mTORC2 activity [124]. [125] showed that the non-steroidal anti-inflammatory drug aspirin-induced the activation of AMPK, leading to the up-regulation of mTORC2-dependent phosphorylation of Akt (S473), which was prevented by knockdown of rictor in hepatoma cell line HepG2 and colon cancer cell line SW480.

The AGC family of kinases, including Akt, PKCα, and SGK1, is composed of substrates of mTORC2 and facilitates mTORC2 modulation of the actin cytoskeletal structure, cell survival, and proliferation [81]. Activation of mTORC2 was found to stimulate phosphorylation of protein kinase C α (PKCα) on S657. In addition, cell survival and proliferation are mediated by mTORC2 via priming Akt for activation through phosphorylation on S473, leading to full activation of Akt by phosphorylation on T308 by PDK1 [127]. Studies have demonstrated that depletion of rictor, mLST8, or mSIN1 of mTORC2 resulted in the ablation of the Akt phosphorylation on S473

[131] investigated the involvement of mTORC2 in glycolysis using mice with liver-specific knockout of rictor (LiRiKO mice). Livers from the LiRiKO mice showed a significant reduction in Akt phosphorylation on S473 and T450, both mTORC2 phosphorylation sites, but the loss of rictor did not affect the phosphorylation of T308, the PDK phosphorylation site. By introducing a constitutively active form of Akt, the effects caused by the loss of rictor were reversed, indicating that mTORC2 mediated glycolysis in an Akt-dependent manner. [132] examined the role of mTORC2 in glycolysis in glioblastoma (GBM) through c-Myc, a critical regulator of cancer cell metabolism.

Both mTORC1 and mTORC2 are involved in innate and adaptive immunity. The mTOR pathways are essential for many immune functions that include suppression of IL-12 and IL-23 production, enhancing M2 macrophage polarization, antigen presentation, and innate immune cell migration. Activation of mTOR in immune cells and other cell types within the tumor microenvironment also affects cancer progression through supporting angiogenesis, metastasis, and drug resistance. The immunological roles of mTOR signaling and its involvement in the tumor microenvironment will not be discussed in this review but have been reviewed in detail elsewhere [134,135].

Rapamycin (sirolimus) is the first mTOR inhibitor discovered as a naturally occurring compound purified from the bacterium Steptomyces hygroscopicus. Rapamycin and its analogs, or rapalogs, inhibit mTORC1 kinase activity by binding to the small mTOR-binding protein FK506-binding protein 12 (FKBP12), and then irreversibly binding to the FRB domain of mTOR, thereby inhibiting the kinase activity of the adjacent catalytic domain [136,137]. In contrast to mTORC1, the mTOR subunit of mTORC2 is insensitive to rapamycin; however, prolonged treatment can disrupt mTORC2 assembly in certain cell types whereby the mTOR protein is unavailable for assembly into mTORC2 as it is sequestered in a complex with rapamycin [138] (reviewed in [139]).

One of the more serious issues with rapamycin and rapalogs is the induction of Akt and ERK signaling in cancer cells that is caused by activation of mTORC2 due to mTORC1 inhibition, as discussed previously [111,147] (for further details, see review [148]). Rapamycin treatment of breast cancer cell lines Akt activation in cancer cells is an unfavorable effect of mTORC1 inhibition as Akt promotes survival and proliferation [147]. Hence, mTORC1 inhibition stimulating Akt and ERK signaling is an unfavorable effect in cancer therapy as this promotes tumor survival and proliferation [111].

Furthermore, AZD8055 treatment impaired cell proliferation of several cancer cell lines and impaired tumor growth in xenograft mouse models [150]. The combination of both AZD2014 and fulvestrant was found to be more effective in inhibiting tumor growth than either drug alone, and patients receiving this combination treatment presented with low toxicities [152]. Although TKIs are an improvement compared to rapamycin and its analogs, they have shown a minimal effect in reducing lung tumor growth in mice with mutant K-Ras [153], and there is greater toxicity associated with these drugs [154]. In phase II clinical trials using patients with non-pancreatic neuroendocrine tumors, CC-223 therapy achieved a median PFS of 19.5 months, an ORR of 7.3%, and a disease control rate of 90.2%, and a tumor size reduction of any magnitude was observed in 73.2% of patients [157].

Furthermore, NVP-BEZ235 has been shown to suppress the growth of hypopharyngeal squamous cell carcinoma (HSCC) cell line FaDu in vitro and in xenograft mouse models. However, clinical trials observed that NVP-BEZ235 therapy was associated with high toxicity and little to no clinical improvement, leading to discontinuation of the treatment [159,160,161]. PI-103 is an inhibitor of mTORC1, mTORC2, DNA-PK, and several PI3K isoforms. Thus far, PI-103 was demonstrated to inhibit the proliferation of various cancer cell lines and tumor growth in xenograft mouse models [162,163].

In the BOLERO-1 phase III clinical trial [165,166], everolimus, trastuzumab, and paclitaxel combination therapy for HER2-positive breast cancer showed an objective response, and median PFS was similar between patients who received the addition of everolimus to the trastuzumab and paclitaxel therapy compared to the addition of the placebo. In the BOLERO-3 phase III clinical trial [167], everolimus, trastuzumab, and mitotic inhibitor vinorelbine combination therapy of patients with trastuzumab-resistant, HER2-positive breast cancer showed minor improvements in response and median PFS. In the LCCC 1025 phase II clinical trial [169], everolimus, trastuzumab, and vinorelbine combined therapy for HER2-positive breast cancer patients with brain metastases showed similar clinical benefit rates relative to other clinical studies using everolimus for HER2-positive breast cancer therapy. Together, these clinical trials observed that combining rapalogs and trastuzumab presented a slight improvement in clinical benefits compared to HER2 inhibition alone.

For the dual PI3K-mTOR inhibitors, clinical trials assessing the efficacy of NVP-BEZ235 therapy for prostate cancer [159], renal cell carcinoma [160], and solid tumors [161] observed high toxicity and no improvement in clinical responses. More recently, the maximum tolerated dose of NVP-BEZ235, as well as formulations and dosage forms, were assessed in patients with solid tumors, including those with HER2-positive breast cancer [173]. The authors noted that the onset of the adverse effects occurred shortly after dosing and may be caused by low absorption and precipitation of the drug at high doses rather than mechanism-based toxicities. Furthermore, certain toxicities associated with PI3K inhibition (hyperglycemia and rash) and mTOR inhibition (pneumonitis) were not observed [173].

Genetically engineered mouse models of HER2-positive breast cancer overexpress Neu/HER2 (wild-type or mutant Neu) in mammary glands under the Mouse Mammary Tumor Virus (MMTV) promoter. MMTV-neu mice constitutively express Neu/HER2 and produce rapidly growing, highly metastatic mammary tumors [177] (reviewed in [178]). Later, in 2008, MMTV-NIC (neu-IRES-Cre) mice were generated, which simultaneously expresses neu and cre recombinase (activated Neu/HER2-MMTV-Cre) under the endogenousErbb2promoter. NIC mice produce aggressive HER2-positive mammary tumors at around 146 days old [179].

When human breast cancer tissue microarrays were analyzed for expression of LKB1, a critical negative regulator of mTORC1, 31% of HER2-positive breast cancers were deficient in LKB1 expression [77]. Furthermore, tumors fromLkb1−/−NIC mice showed enhanced phosphorylation of the S6K1 substrate ribosomal protein (S6), elevated ATP levels, and changes in metabolic enzymes and metabolites indicative of mTORC1 hyperactivation [77]. [180] assessed the role of LKB1 in breast cancer, including HER2-positive breast cancers, using immunohistochemical analysis of tumors from early breast cancer patients and in silico analysis obtained from the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) dataset. Although LKB1 protein expression did not correlate with HER2 status, high LKB1 expression in HER2-positive breast cancer patients correlated with improved overall survival, consistent with the findings of Andrade-Vieira et al.

Using the mammary epithelial cells (MECs) of tumors isolated from both fromLkb1−/−NIC and NIC mice, treatments using mTOR inhibitors (rapamycin, Torin-1, and AZD8055) reduced phosphorylation of S6; however, Torin-1 and AZD8055 reduced the levels of phosphorylated Akt on both S473 and T308 When considering mitochondrial function, AZD8055 treatment reduced the mitochondrial content of primary mammary tumors isolated from Lkb1−/−NIC mice, but not in mammary epithelial cells isolated from control wild-type female littermates. Tumors isolated from AZD8055-treatedLkb1−/−NIC mice showed significantly reduced expression of glycolytic enzymes (hexokinase 2, lactate dehydrogenase (LDH), and pyruvate dehydrogenase (PDH)) and phosphorylation of S6 compared with the vehicle control. However, tumors from AZD8055-treated mice also showed strong induction of ERK and p90RSK phosphorylation

4. Glycolysis in Breast Cancer

Glycolysis uses glucose to generate two molecules of pyruvate and energy in the form of ATP [182] (reviewed in [183]). Under aerobic conditions, pyruvate is transported into the mitochondria and converted to citrate and CO2. When oxygen is limited, pyruvate is metabolized via anaerobic glycolysis, which generates ATP less efficiently but 100X more rapidly compared to OXPHOS [185]. In anaerobic glycolysis, LDH catalyzes the reduction of pyruvate and regeneration of NAD+, where pyruvate and NADH are converted to lactate, NAD+, and two ATP molecules.

[186] made the landmark observation that tumor cells metabolized high levels of glucose to produce ATP and lactate in the presence of oxygen. The metabolic shift of OXPHOS to aerobic glycolysis in tumor cells is known as the “Warburg effect”. Interestingly, lung tumor cells isolated from one of the NSCLC mouse models relied more on glutamine metabolism to support cell growth, whereas tumor cells analyzed directly from the same NSCLC mouse model did not rely on glutamine metabolism. The observation of metabolic differences between lung cancer cells in vitro and in vivo suggested a role of the tumor microenvironment in influencing cancer cell metabolism.

Several studies have observed that resistance to targeted therapies in cancer cells is associated with increased glycolytic activity and expression of glycolytic enzymes [195,196,197]. The concept of metabolic reprogramming has gained popularity as a means for tumors to adapt to the metabolic requirements for survival (for further details, please refer to reviews [198,199]).

HER2 overexpression in breast cancer cell lines increased glycolysis as indicated by increased glucose uptake and lactate production, and decreased oxygen consumption rates [200]. [200] found that HER2 overexpression in breast cancer cell lines Re-expression of ZBTB1 in HER2-expressing breast cancer cell lines reduced lactate production, glucose uptake, and down-regulated LDH and HK expression, indicating that the elevated expression of HER2 due to the loss of ZBTB1 promotes aerobic glycolysis [195]. The glycolytic activities in HER2-positive breast cancer cell lines are reduced in response to trastuzumab treatment.

As the HER2-E subtype expresses higher levels of HER2 and glycolytic metabolites compared with other HER2-positive breast cancer subtypes, the HER2-E subtype also displayed higher levels of phosphorylated S6K, a substrate of mTORC1 [208]. Inhibition of mTORC1/mTORC2 (Torin-1, AZD8055) or mTORC1 (rapamycin) down-regulated LDH expression and had little to no effect on PDH expression in the primary tumor cells isolated fromLkb1−/−NIC mice compared to the cells from NIC mice [77,151]. Characterization of mitochondrial content, size, and cristae density were greater in mammary tumors fromLkb1−/−NIC mice compared with mammary glands from control WT mice. This study demonstrates that therapies that simultaneously target mTORC1/mTORC2 and glycolytic metabolism in cancer produce the best therapeutic outcome against HER2-positive breast cancer.

Tumor cells can develop a dependency on glycolysis for survival. Glucose analogs cause glucose deprivation, resulting in the suppression of glycolysis as they cannot be metabolized by cells. 2-deoxy-D-glucose (2-DG) is a glucose analog that is taken into the cytosol through glucose transporters (GLUTs), where hexokinase phosphorylates 2-DG to generate 2-DG-P; however, phosphohexose isomerase is not able to metabolize 2-DG-P any further (reviewed in [209]). Here, downstream glycolysis and production of cellular ATP are inhibited by the accumulation of 2-DG, which is associated with impaired cell cycle progression and enhanced cell death of tumor cells [210].

However, antiproliferative and cell death-promoting effects of 2-DG have been observed in vitro and in vivo in cancer cells [213]. Treatment using 2-DG induces endoplasmic reticulum stress, leading to autophagy. This results from the accumulation of misfolded proteins in the ER lumen concomitant with ER stress and the unfolded protein response, a mechanism of inhibiting protein translation to relieve ER stress [213]. Inhibition of autophagy prevented 2-DG-induced autophagy and ER stress but did not reverse the depletion of ATP.

Suppression of glycolysis in HER2-positive breast cancer has been observed to reduce HER2-driven mammary tumor cell growth in vitro as well as in vivo with mouse models [214]. In the Lkb1−/−NIC mouse model of HER2-positive mammary cancer, 2-DG monotherapy reduced tumor burden and growth compared with vehicle-treated mice, but to a lesser extent than AZD8055 monotherapy [151]. Tumors from 2-DG-treated mice showed reduced glycolysis, oxygen consumption rate, mitochondrial content, and down-regulation of HK, PDH, and LDH expression. Interestingly, 2-DG induced phosphorylation of AMPK (T172) concomitant with reduced mTORC1 activity as observed from reduced phosphorylation of mTOR (S2448) and S6K1 (T389) in various cell types.

Studies assessing the efficacy of 2-DG and other metabolic interventions in combination with various anticancer therapies have observed tolerable adverse effects from the addition of 2-DG [219] (reviewed in [220]). Preclinical studies determined that high dosages negatively impacted the respiratory frequency and mean arterial blood pressure [221], and reversible cardiac toxicity has been associated with 2-DG treatment in rats [222]. Despite the therapeutic potential of 2-DG for cancer treatment, studies have shown little to no effect of 2-DG treatment alone in inhibiting tumor growth in preclinical mice models, including genetic [151] and xenograft models [223]. In a clinical trial conducted in patients with a variety of solid tumors, Raez et al.

Consistent with this, our laboratory observed an up-regulation of ERK phosphorylation, concomitant with a decrease in glycolytic activity and tumor growth, in mammary tumors As AZD8055 did not completely block glycolytic activity in mammary tumors ofLkb1−/−NIC mice, combination treatment was performed by adding 2-DG to the AZD8055 treatment to further inhibit glycolysis in addition to mTORC1/2. These studies demonstrate the benefit of combining glycolytic inhibition via 2-DG with mTORC1/2 inhibition, in which the addition of 2-DG prevents the mTOR inhibitor-dependent induction of pro-survival MAPK pathway signaling. These studies strongly suggest that the addition of 2-DG may improve the efficacy of therapies using mTOR inhibition, as well as other targeted therapies, by mitigating factors that contribute to drug resistance such as activation of pro-survival signaling and drug efflux.

This entry is adapted from the peer-reviewed paper 10.3390/cancers13122922

This entry is offline, you can click here to edit this entry!