Targeting Breast Cancer: Comparison
Please note this is a comparison between Version 1 by Hamidreza Montazeri Aliabadi and Version 2 by Camila Xu.

Breast cancer became the most diagnosed cancer in the world in 2020. Chemotherapy is still the leading clinical strategy in breast cancer treatment, followed by hormone therapy (mostly used in hormone receptor-positive types).

  • breast cancer
  • targets
  • signaling pathway
  • druggable
  • therapy

1. Introduction

Cancer is still among the leading causes of morbidity worldwide. In 2020, more than 19 million new cases of cancer were diagnosed, and almost 10 million cancer-caused deaths were reported [1]. In 2020, breast cancer became the most diagnosed cancer in the world, with over 2.3 million new cases and 685,000 in this year [2]. This grim statistic is partly due to the drop in deaths caused by infectious diseases (better sanitation, new antibiotics, and vaccine development) and better control and prevention of cardiovascular diseases [3].
The innate resistance of unresponsive cells is usually explained by tumor heterogeneity. In 2015, Sottoriva et al. proposed a ‘Big Bang’ model of tumor initiation that suggests that after initial oncogenic mutations, future generations acquire further mutations, which are present in discrete populations of cells, leading to spatial heterogeneity [4]. An even more diverse pattern has been observed in other types of cancer. Amir et al. studied two human acute lymphoblastic leukemia samples with viSNE technology and reported a large, irregular population of cells that were more different than similar [5]. Personalized medicine (based on the biomarker expression in each specific patient) has been suggested as a solution to the limited response rate in naïve cells. However, this intra-tumor heterogeneity remains a major hindrance. The sub-population with intrinsic resistance to therapeutic assault would survive and outgrow other cells due to the selection pressure, which promotes relapse and results in an abundance of cells that were once minorities [6]. This “Darwinian clone selection” has been well documented in different types of cancer in response to a variety of molecularly targeted drugs [7].
Efflux proteins (e.g., P-glycoprotein) and their role in acquired resistance to chemotherapeutic agents have been well studied. This desensitization of originally responsive cells has also been observed in response to molecularly targeted drugs [8][9][8,9]. This acquired resistance has been reported in different types of cancer (e.g., non-small-cell lung cancer [10][11][10,11]) and for different targets (e.g., hormonal therapy in estrogen receptor-positive breast cancer [12] or receptor tyrosine kinases [13][14][13,14]). While point mutations are widely accepted mechanisms for this resistance development [15][16][17][18][19][15,16,17,18,19], the ability of cancer cells to “switch” to an alternative protein for their survival (known as plasticity) is another important factor. In this case, the initial response to the molecularly targeted drug (as the result of inhibition of the targeted protein) might be diminished due to the overactivation of other proteins and/or signaling axes that could compensate for the loss of function of the inhibited protein [20][21][20,21].

2. The Familiar

2.1. Hormone Therapy

Tamoxifen was first synthesized as a contraceptive in 1962. The project failed since this molecule surprisingly stimulated ovulation. In the 1980s, clinical trials showed its efficiency in breast cancer when used with chemotherapy. Consequent trials showed the efficiency of tamoxifen in the prevention of the development or re-appearance of breast cancer, and a new approach to breast cancer therapy was born [22][29]. Hormone therapy still takes up a big share of the FDA-approved drugs for breast cancer therapy, and it is not surprising since the majority of the diagnosed breast cancers are estrogen receptor (ER) positive (~80%) or ER and progesterone receptor (PR) positive (~65%) [23][24][30,31]. Among eight FDA-approved drugs categorized as hormone therapy, there are three aromatase inhibitors that lower estrogen levels, three selective estrogen receptor modulators (SERMs), one elective estrogen receptor downregulator (SERD; fulvestrant) that destroys the receptor, and one luteinizing hormone-releasing hormone (LHRH) agonist that, with chronic use, reduces estradiol levels. Steroid hormones play a role in carcinogenesis in breast cancer and would result in enhanced cell growth, development, differentiation, and homeostasis [25][32]. However, there is much more to the estrogen/progesterone receptors and their signaling than meets the eye at first glance. The effect of estrogen is mainly exerted through estrogen receptor alpha (ERa) and/or estrogen receptor beta (ERb) [26][33]. Inactive Era binds to heat-shock proteins in the cytoplasm. In the presence of estrogen, it dissociates and binds to estradiol, dimerizes, enters the nucleus, and binds to estrogen response element (ERE) to act as a transcription factor to enhance expression of Cyclin D1, which activates CDK4/6 [27][34]. It also increases the expression of the mouse double minute 2 homolog (MDM2), which promotes cell survival via different mechanisms, including suppression of p53 [28][35]. However, estrogen and progesterone signaling pathways have a non-genomic pathway as well, which includes crosstalk with growth factor receptors and G-protein-coupled (GPR) receptors [29][36]. It has been reported that estrogen transactivates EGFR via GPR30, which in turn activates the Ras/Raf pathway and its downstream effectors (MEK and ERK-1/-2) [30][37]. It has also been shown that PR could activate Src-MAPK and AKT pathways [31][38].

2.2. HER Family of Receptors

Targeting the family of HER receptors (especially HER1, EGFR, and HER2) seems like a logical approach in HER2-positive breast cancer, which makes up approximately 30% of breast cancer cases [32][39]. However, the signaling cascades that are triggered by this family of receptor tyrosine kinases (RTKs) involve a wide array of proteins and intracellular mechanisms, including the same mechanisms that are activated by estrogen. The function of HER2 is dependent on dimerization with another HER2 (homodimerization) or other members of the family (heterodimerization) [33][40]. This dimerization is the trigger for the activation of a variety of proteins, enzymes, messengers, and transcription factors that induce an array of responses involved in the carcinogenesis of the mammary gland [34][41]. Unlike other members of the receptor family, HER2 does not have a ligand ([35][42]) and is reported to stabilize and enhance dimerization [34][41]. The dimerization mainly activates the Ras/Raf/MEK/ERK and PI3K/AKT pathways, which increase the expression of Cyclin D1 and activate CDK4/6 directly and indirectly, respectively [35][36][37][42,43,44]. There is also evidence for activation of the JAK/STAT pathway by HER2 and EGFR homo/hetero dimers, which would lead to enhanced expression of proteins that would activate epithelial-to-mesenchymal transition (EMT), which increases the risk of metastasis [38][39][45,46].

2.3. CDK4/6

As mentioned before, three drugs targeting CDK4/6 have been approved by the FDA to be used in HR-positive and HER2-negative breast cancers. CDK4 and CDK6 are members of a family of serine/threonine kinases that are activated by binding to cyclins (mostly cyclin D in breast cancer) and enhance cell cycle progression and cell proliferation [44][54]. The CDK4/6 complex is an important mediator for transition into S phase and plays an important role in carcinogenesis and the progression of breast cancer [45][55]. On the other hand, resistance to HER2 targeting molecules has encouraged studies of the effect of CDK4/6 inhibitors in HER2-positive cases that show resistance to HER2 inhibitors [46][47][56,57]. It is important to note that CDK4/6 is considered a downstream effector for both estrogen- and RTK-triggered pathways. Many additional first- and second-generation CDK4/6 inhibitors are in clinical trials and have been reviewed recently in multiple publications [48][49][50][58,59,60]. Despite promising results, resistance to CDK4/6 inhibitors also seems to be inevitable and seems to involve a variety of mechanisms, including abnormal activation of CDK4/6, loss of retinoblastoma protein (pRb), cyclin E activation, loss of PTEN, and activation of alternative pathways including Ras, fibroblast growth factor receptor 1 (FGFR1), and/or PI3K/AKT pathways [51][52][61,62].

3. The Emerging

3.1. Immune Checkpoint Inhibitors

Inherent genetic and epigenetic changes in cancer cells create a wide variety of antigens, and therefore, tumor cells develop immune resistance mechanisms, including immune checkpoints [53][63]. Immunotherapy has been a significant recent advancement in cancer treatment, and while breast cancer did not initially seem to be a great candidate for this approach (due to low immunogenicity), immune checkpoint inhibitors have found their way into breast cancer therapy [54][64]. Inhibition of immune checkpoints can be achieved via two general approaches: targeting the ligand expressed on the cancer cell or the inhibitory receptor on the cancer-specific T cells. Both approaches have been investigated, and each presents different opportunities and challenges. Among the targets in this category, the PD-1/PD-L1 pair has been explored more extensively, and both FDA-approved drugs in this category for breast cancer therapy target this pair. The other pairs that trigger an inhibitory signal for immune response to cancer cells are CD80/CTLA-4, MHC/LAG3, Gal9/TIM3, and PVR/TIGIT.
Table 12.
Summary of ongoing or completed clinical trials involving immune checkpoint inhibitors in breast cancer.
Video Production Service