Skip to main content
Cell Proliferation logoLink to Cell Proliferation
. 2016 Jun 16;49(4):409–420. doi: 10.1111/cpr.12266

Cell‐specific biomarkers and targeted biopharmaceuticals for breast cancer treatment

Mei Liu 1, Zhiyang Li 1,2, Jingjing Yang 3, Yanyun Jiang 3, Zhongsi Chen 1, Zeeshan Ali 3, Nongyue He 1,, Zhifei Wang 3,
PMCID: PMC6496337  PMID: 27312135

Abstract

Breast cancer is the second leading cause of cancer death among women, and its related treatment has been attracting significant attention over the past decades. Among the various treatments, targeted therapy has shown great promise as a precision treatment, by binding to cancer cell‐specific biomarkers. So far, great achievements have been made in targeted therapy of breast cancer. In this review, we first discuss cell‐specific biomarkers, which are not only useful for classification of breast cancer subtyping but also can be utilized as goals for targeted therapy. Then, the innovative and generic‐targeted biopharmaceuticals for breast cancer, including monoclonal antibodies, non‐antibody proteins and small molecule drugs, are reviewed. Finally, we provide our outlook on future developments of biopharmaceuticals, and provide solutions to problems in this field.

1. Introduction

Breast cancer is the most common cancer among women in the world, which is reported to be a heterogeneous tumour with well‐defined molecular subtypes.1 In 2015, an estimated 231 840 new cases of invasive breast cancer were diagnosed in women in the United States, along with 40 290 new cases of breast cancer death.2 In China, breast cancer has also become a major health problem of women especially those living in urban areas with an overall morbidity of 24.20 per 100 000.3, 4, 5, 6 Furthermore, the incidence of breast cancer is unfortunately increasing. Despite the extraordinary progress in understanding the molecular mechanisms underlying carcinogenesis, tumour promotion, and mammographic screening for the early detection and treatment of breast cancer, the global mortality of breast cancer patients is still unacceptably high, and the 5‐year survival rate of metastatic breast cancer (stage IV) is below 15%.5

So far, the common breast cancer treatment is surgery supplemented with radiation therapy, various forms of systemic adjuvant chemotherapy, and hormonal therapy, which may also kill the normal cells as a side effect. Targeted therapy has shown great promise as a precision cancer therapy by binding to cancer cell‐specific biomarkers. In comparison with the traditional medicine, the targeted therapy for breast cancer seems to be more effective and lower toxic. For the development of such targeted therapy, it is necessary to investigate the biology of breast cancer cells and especially understand the cell‐specific biomarkers expression and tumour subtyping in advance. Nowadays, many efforts have been made to study the genes, proteins, metabolites, and signalling pathways, which are related to the growth and metastasis of breast cancer.7 Several cell‐specific biomarkers of breast cancer cells, such as oestrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor receptor 2 (HER2), vascular endothelial growth factor (VEGF), and Ki‐67, have been thus discovered.8, 9, 10, 11 These cell‐specific biomarkers can not only be utilized as targets for novel drugs delivery but also be used for early detection and classification of breast cancer, which will greatly accelerate the development of the targeted therapies for breast cancer.8, 9, 10, 11

A biopharmaceutical, also known as a biological medical product, biological or biologic, is any medicinal product obtained from biological sources. Different from chemically synthesized pharmaceuticals, they possess high affinity, specificity, selectivity, low toxicity as well as perfect efficacy. Since the first approval of trastuzumab by the United States Food and Drug Administration (FDA) in 1998, several biopharmaceuticals, including monoclonal antibodies (mAbs), non‐antibody proteins and other small molecules, have been extensively developed to target specific antigens during cancer progression.12, 13, 14, 15, 16, 17, 18, 19 Table 1 lists the innovative biopharmaceuticals currently approved by the FDA and the European Medicine Evaluation Agency for the treatment of breast cancer.

Table 1.

The currently approved and emerging biopharmaceuticals for breast cancer targeted therapy

Name Target Type Mechanism of action References
Trastuzumab HER2 Humanized IgG1 Triggers HER2 internalization and degradation, and inhibits HER2‐related cell signalling; ADCC. 20, 21, 22, 23, 24, 25
Pertuzumab HER2 Humanized IgG1 Inhibits the heterodimerization of HER2 with other HER family, and hinders HER2‐related cell signalling; ADCC. 26, 27, 28, 29, 30
T‐DM1 HER2 Humanized IgG1 Targets intracellular delivery of cytotoxic emtansine; Triggers HER2 internalization and degradation; Inhibits HER2‐related cell signalling. 31, 32, 33, 34, 35
Denosumab RANKL Humanized IgG2 Inhibits the receptor RANK on osteoclasts and blocks the downstream signalling pathways of breast cancer‐mediated bone diseases. 36, 37, 38
Ertumaxomab HER2 and CD3 Murine and rat biospecific IgG2 ADCC and ADCP 39, 40, 41
Bevacizumab VEGF Humanized IgG1 Inhibits VEGF‐mediated angiogenesis 42, 43, 44, 45, 46
Ramucirumab VEGFR2 Humanized IgG1 Inhibits VEGFR2‐mediated angiogenesis 47, 48
Aflibercept VEGF Recombinant peptide‐antibody fusion protein Inhibits VEGF‐mediated angiogenesis 49, 50, 51, 52

T‐DM1: Ado‐trastuzumab emtansine; RANKL: Receptor activator of NF‐kB ligand; TKI: Tyrosine kinase inhibitor; ADCC: Antibody‐ dependent cell‐mediated cytotoxicity; ADCP: Antibody‐dependent cellular phagocytosis.

In the past decades, targeted therapy of breast cancer was one of the hottest areas of cancer research, and hundreds of articles, related to cell‐specific biomarkers or therapies, have been published. Although rich and colourful reviews are available on both cell‐specific biomarkers and correlated biopharmaceuticals for the targeted therapy of breast cancer, they have been seldom discussed together as one topic. To better understand the development of targeted therapy in breast cancer, in this article, we first discuss the cell‐specific biomarkers for biopharmaceuticals targeting, then review the innovative and generic‐targeted biopharmaceuticals for breast cancer treatment, and finally make an outlook of the future development of targeted biopharmaceuticals for breast cancer as well as provide the solutions to the problems in this field.

2. Cell‐specific biomarkers for breast cancer subtyping and targeted biopharmaceuticals delivery

Cell‐specific biomarkers refer to specific molecules that are expressed or overexpressed on particular cancer cells, which provide the foundation of breast cancer detection, classification, risk assessment, and treatment response prediction and also play an important role in targeted approaches in both primary and metastatic breast cancer therapy.53 In order to achieve personalized therapy, prior knowledge of breast cancer subtyping based on cell‐specific biomarkers is required. Currently, the methods for the determination of cell‐specific biomarkers mainly consist of immunohistochemistry, enzyme‐linked immunosorbent assay, polymerase chain reaction and fluorescence in situ hybridization.54, 55, 56, 57, 58, 59, 60, 61, 62 After years of research and testing, several cell‐specific biomarkers, including ER, PR and HER2, have been discovered and further used as gold standard biomarkers for both breast cancer classification and targeted therapy. Based on above three cell‐specific biomarkers, breast cancer can be classified into four major intrinsic subtypes: Luminal A, Luminal B, HER2‐overexpressed and basal‐like.53, 63, 64, 65, 66 The treatments corresponding to four different breast cancer molecular subtypes are showed in the Table 2. Additionally, VEGF, which plays a very important role in cancer angiogenesis, can also be regarded as a cell‐specific target for breast cancer treatment. However, owing to the molecular heterogeneity and considerable histopathology, the molecular fingerprints for breast cancer classification are still unaccomplished, and new cell‐specific tumour biomarkers for both breast cancer classification and targeted therapy need to be discovered.

Table 2.

Immunohistochemical criteria for breast cancer intrinsic subtyping and corresponding treatment 53, 63, 64, 65, 66

Subtype ER PR HER2 Treatment
Luminal A Either ER or PR positive Negative Endocrine therapy
Luminal B Either ER or PR positive Positive Endocrine therapy + anti‐HER2 therapy + Chemotherapy
HER2 overexpressed Negative Negative Positive Anti‐HER2 therapy + Chemotherapy
Basal‐like Negative Negative Negative Chemotherapy

ER: Oestrogen receptor; PR: Progesterone receptor; HER2: Human epidermal growth factor receptor 2.

2.1. ER and PR

Oestrogen receptor and PR are expressed in the cell nuclei of about 60%–70% breast cancer cells.63, 64, 65 ER, which was first identified in the late 1960s, plays an important role in breast cancer progression.67 ER‐positive breast cancer is thought to be the best variant, with effective strategies to block, downgrade or deprive the ER of its efficacy, when compared with other subtypes.68 As a nuclear transcription factor, ER participates at the regulation of transcriptional activity of breast tissues, which is termed nuclear‐initiated steroid signalling. When ER interacts with oestrogen, they will form a co‐regulator complex, and regulate transcription of specific genes, such as PR, TFF1, GREB1 and PDZK1.68, 69, 70 There are two isoforms of ER: ERα and ERβ. Normally, the expression of ERα is essential for the development of normal mammary gland, while a prominent expression is often observed in breast carcinoma sites.67, 68 ERβ is highly homologous to ERα both in the DNA‐binding domain and ligand‐binding domain. However, for both ERα‐positive breast cancer and ERα‐negative one, ERβ can be observed, which plays a key role in breast cancer classification and endocrine therapy.67

Progesterone receptor, which regulates ER expression, is another important hormone receptor biomarker in breast cancer classification and targeted therapy. Similar to ER, it also exists in two kinds of forms: PRA and PRB.67 PR plays a very important role in the lobuloalveolar development of breast. However, an overexpression of PRA always means the elevated risk of breast cancer.68 So PR is usually used as an indicator of nuclear ER pathway and tumour aggression and serves as an instruction for endocrine therapy.71 Furthermore, breast cancer with a remarkable overexpression of PR is always less aggressive compared with PR‐negative breast cancer.68, 71

2.2. HER2

Human epidermal growth factor receptor 2 is a 185 kDa transmembrane protein belonging to the ErbB family of receptor tyrosine kinases.72, 73, 74, 75 It can regulate a series of cellular processes such as cell proliferation, differentiation and survival.21, 76 As a perfect predictive biomarker associated with anti‐HER2 biopharmaceutical targeted therapies, HER2 protein is overexpressed in approximately 20%–25% of all invasive breast cancer patients.21, 77, 78 Consequently, a couple of anti‐HER2 biopharmaceuticals, such as trastuzumab,20, 79 pertuzumab28 and ado‐trastuzumab emtansine (T‐DM1), have been developed and commercialized.26, 80 The detail mechanisms of these HER2‐targeted biopharmaceuticals are displayed in Fig. 1.26 Moreover, it is noted that the overexpression of HER2 protein is much due to gene mutation of HER2, which happens in about 1%–2% of breast cancer.81

Figure 1.

Figure 1

Molecular mechanisms of novel biopharmaceuticals for HER2‐targeted therapy. Reprinted with permission from reference 26. Copyright@ Nature Publishing Group

Due to the fact that HER2 status is an important biomarker in guiding personalized HER2 therapy, it is of significance to accurately determine its status in certain breast cancer tissues. In clinical treatment, HER2 targeted therapy should be recommended only when the HER2 test result is positive. The HER2‐positive criteria by immunohistochemistry is 3+ based on complete circumferential membrane staining or the HER2/CEP17 ratio ≥2.0 by fluorescence in situ hybridization.82 However, these two techniques can give false‐positive or false‐negative results to some extent. Therefore, new methods for clinical evaluation of HER2 expression should be developed.

2.3. VEGF pathways

Vascular endothelial growth factor and its correlated signalling pathways play key roles in tumour angiogenesis.44 The VEGF family consists of several polypeptides, namely VEGF‐A, VEGF‐B, VEGF‐C, VEGF‐D and PlGF, among which VEGF‐A is not only an endothelial growth factor but also a regulator of vascular permeability.44, 83, 84 VEGF‐A always interacts with VEGFR1 or VEGFR2, two transmembrane receptor tyrosine kinases (RTKs), to promote endothelial cells migration and mitosis and withstand their apoptosis or senescence.84, 85, 86 The detail mechanisms of VEGF‐mediated signalling pathways are as follows: when VEGF‐A interacts with VEGFR2, the VEGFR2 PTKs site will autophosphorylate, which produces inositol 3,4,5‐trisphosphate and diacylglycerol. They are two kinds of second messenger molecules that can activate the protein kinase C and the extracellular signal‐regulated kinase pathway, which eventually result in endothelial cells multiplication, migration and invasion.85, 86, 87 VEGF‐mediated angiogenesis can not only affect reproduction, embryogenesis and skeletal growth in the physiology conditions but also seriously influence pathological angiogenesis. VEGF‐targeted biopharmaceuticals are thus demonstrated to decrease tumour capillary density and suppress tumour growth by specifically inhibiting VEGF‐mediated signal transduction pathway.88, 89, 90, 91

Due to the fact that angiogenesis plays a very important role in the progression of breast cancer, the development of VEGF‐targeted biopharmaceuticals has been attracting significant attention from scientists and clinic doctors. Currently, there are a couple of VEGF‐targeted biopharmaceuticals under investigation for breast cancer treatment, including bevacizumab,92 ramucirumab93 and aflibercept.86, 94 The detail mechanisms of such VEGF‐targeted biopharmaceuticals are illustrated in Fig. 2.86 Moreover, other potential VEGF‐targeted biopharmaceuticals for breast cancer are also being investigated, and more tests need to be done to evaluate their effects. Most recently, the potential synergistic therapies between VEGF‐targeted biopharmaceuticals‐based therapy and other forms of treatment are in full swing, which holds new promise for breast cancer therapy and prognosis.

Figure 2.

Figure 2

Schematic of novel anti‐angiogenesis biopharmaceuticals targeting VEGF pathways in breast cancer. Reprinted with permission from reference 86. Copyright@ Informa Healthcare

2.4. Other cell‐specific biomarkers for breast cancer targeted therapy

Besides the above biomarkers and pathways, other biomarkers, such as HER3,95 MUC196 and insulin‐like growth factor receptor,97 also have the potential for breast cancer targeted therapy. However, up to now, few biopharmaceuticals based on such biomarkers have been developed.

3. Targeted biopharmaceuticals for breast cancer treatment

3.1. Monoclonal antibodies against HER2

3.1.1. Trastuzumab

Trastuzumab, used as the first‐line treatment for HER2‐positive metastatic breast cancer, is a fully humanized version of a murine HER2‐targeted IgG1 monoclonal antibody with a molecular mass of 148 400 Da.98, 99, 100, 101 Since it was first approved by FDA in 1998, trastuzumab has achieved great clinical success and saved thousands of lives of breast cancer patients. The functions of trastuzumab for breast cancer treatment mainly involve HER2 monoubiquitination, anti‐angiogenic properties, HER2 cleavage and extracellular domain shedding, DNA repair inhibition and antibody‐dependent cell‐mediated cytotoxicity (ADCC).102, 103, 104, 105, 106

Large amounts of clinical trials indicate that trastuzumab is well tolerated and can work effectively either in monotherapy or in combination with other cytotoxic agents on HER2‐positive metastatic breast cancer. However, the patients initially treated with trastuzumab would suffer from an infusion reaction of either fevers or chills, which may be an obstacle to its application.106, 107, 108, 109 Additionally, the development of the related drug resistance in patients would make HER2‐positive breast cancer more difficult to cure. The proposed mechanisms of trastuzumab resistance mainly include the prevention of antibody‐receptor interaction, phosphatase deficiency, inhibition of cell proliferation, activation of PI3K/AKT pathway, damages of insulin‐like growth factor‐1 receptor (IGF‐1R) signalling pathways, and sheltering of HER2 receptor or epitope by mucin 4 (MUC4).96, 110, 111, 112, 113, 114, 115, 116 Meanwhile, both the upregulation of HER2 expression and cyclin E overexpression could lead to trastuzumab resistance as well.115, 117 Finally, owing to the fact that the expression of HER2 in cardiac myocytes plays a significant role in both embryogenesis and myocyte repair, trastuzumab can slightly increase the risk of symptomatic cardiac events (about 4% every 4 years),118 which is another shortcoming of trastuzumab in breast cancer treatment.

Nowadays, combination therapy has become the hottest field in tumour treatment.119 At the beginning of trastuzumab treatment, the overall response rate (ORR) of HER2‐positive metastatic breast cancer patients can reach 26%,100, 120 while the combination therapy of trastuzumab with other chemical agents can reach up to approximately 50%.107 The chemical agents currently suggested for the combination therapy of HER2‐positive breast cancer include paclitaxel,121 docetaxel,122, 123 carboplatin,124, 125 capecitabine,126 gemcitabine127, 128 and cisplatin.129

3.1.2. Pertuzumab

Pertuzumab is a humanized IgG1 monoclonal antibody that binds to the extracellular domain II of HER2. It is the new class of HER family (HER1, HER2 and HER3) dimerization and ligand‐activated signalling inhibitor.28, 130 The mechanism of pertuzumab for breast cancer therapy is that pertuzumab can specifically bind with the cysteine‐rich region of HER2 extracellular domain II and sterically block the dimerization of HER2 with HER1, HER3 or HER4, which leads to the inhibition of intracellular signalling pathway and further preventing the proliferation of tumour cells.131, 132, 133, 134

Different from trastuzumab, pertuzumab binds to the domain II of HER2 receptor.135, 136 Therefore, the combined therapy of trastuzumab and pertuzumab will block the HER2‐mediated signal transduction more completely by receptor crosstalk.132, 137 To date, pertuzumab has been assessed both in monotherapy and in combination therapy with trastuzumab for HER2‐positive breast cancer treatment.28, 122, 136, 137, 138, 139 Clinical trials indicate that combination therapy of trastuzumab and pertuzumab synergistically suppresses the growth of HER2‐overexpressed breast cancer cell lines.73, 140 An anti‐tumour activity study of trastuzumab and pertuzumab alone as well as in combination for HER2‐positive breast cancer xenografts was conducted by Scheuer and his colleagues.137 The results indicate that the combination therapy of trastuzumab and pertuzumab shows more robust anti‐tumour effect than each agent applied separately.137, 138 In addition, pertuzumab is also well tolerated and shows very little haematological and cardiac toxicity.

Other strategies of synergy therapy using pertuzumab in combination with other chemical agents have also been investigated.24, 28, 131 For example, the combination therapy of pertuzumab and lapatinib performed a greater tumour suppression effect than the results obtained in monotherapy.141 The efficacy of pertuzumab in combination with trastuzumab and docetaxel in neoadjuvant strategies for HER2‐positive breast cancer treatment was evaluated in a phase II, and the results showed that a combination of three chemicals had better effect in HER2‐positive breast cancer treatment than other dual combination therapies.142 Moreover, a phase III study was conducted to investigate the combination of trastuzumab and docetaxel with or without pertuzumab, and the results indicated that breast cancer patients who received pertuzumab had a greater progression‐free survival (PFS) (12.4 months for control arm vs 18.5 months for pertuzumab arm, HR 0.62; 95% CI, 0.51–0.75; P<.001) and a stronger trend for improved survival.123, 143, 144

3.1.3. Ertumaxomab

Ertumaxomab is a trifunctional bispecific monoclonal antibody binding to HER2/neu and CD3. It specifically binds to Fcγ type I/III receptors to form a tri‐cell complex among tumour cells, T cells and accessory cells, which eventually leads to the tumour cell death.145, 146 It is produced by a quadroma cell line resulted from the fusion of a specific anti‐CD3 rat hybridoma cell line and an anti‐HER2 mouse hybridoma cell line.145 The function of ertumaxomab is related to immune response such as ADCC and antibody‐dependent cellular phagocytosis.145, 147 The combination therapy of ertumaxomab with other chemical drugs has also been studied, which indicates that ertumaxomab is a new promising monoclonal antibody for HER2‐positive breast cancer targeted therapy.148

3.2. Monoclonal antibodies against VEGF

3.2.1. Bevacizumab

Bevacizumab is a recombinant humanized IgG1 monoclonal antibody that binds and devitalizes soluble VEGF‐A molecules.42, 43, 44, 45, 46 It is the first anti‐VEGF drug used for the breast cancer treatment, which shows great promise in breast cancer targeted therapy. Although bevacizumab has been approved by the European Medicines Agency for the first‐line treatment of metastatic breast cancer in 2007,149, 150 there are still many controversies in utilizing bevacizumab for breast cancer treatment due to its adverse effects of hypertension, bleeding events, proteinuria and sensory neuropathy.151, 152 Owing to the failure in demonstrating the clinical benefits of bevacizumab in combined therapy with other chemical agents, in 2011, the FDA banned the use of this drug for metastatic breast cancer treatment after its first approval in 2008.153, 154 Fortunately, the latest research has verified the usefulness of bevacizumab in first‐line standard chemotherapy for metastatic breast cancer treatment.45

Bevacizumab was usually tested in combination with other chemotherapeutic agents or in the neoadjuvant therapy strategies,55, 155, 156, 157 such as docetaxel,158, 159 capecitabine160, 161, 162 and taxanes.163 The results of first‐ and second‐line trials demonstrate positive impact on the endpoints of PFS and ORR of breast cancer patients.164, 165, 166 The newest results of bevacizumab in combination with chemical drugs for breast cancer treatment indicate that there is a great improvement on endpoints of ORR, PFS (8.1 months vs 5.4 months), overall survival rates (39.0 months vs 23.7 months) and pathological complete response vs patients treated with chemical drugs alone.158, 165, 166, 167, 168, 169

3.2.2. Ramucirumab

Ramucirumab is a fully humanized IgG1 monoclonal antibody, which specifically binds to the extracellular domain of vascular endothelial growth factor receptor 2 (VEGFR‐2). It can inhibit endogenous VEGF signalling of tumour angiogenesis.47, 170 The early clinical trials of ramucirumab in various malignant tumours indicate that ramucirumab is well tolerated and produces satisfactory results in suppressing angiogenesis in about 69% of evaluated tumours.149, 171 Similar to bevacizumab, the treatment of ramucirumab also leads to the occurrence of hypertension and venous thrombosis but such adverse effects could be alleviated by taking good care of patients.47, 170 It should be noted that ramucirumab has been approved by FDA as the first monotherapy agent for the treatment of advanced or metastatic gastric cancer or gastroesophageal junction adenocarcinoma patients who had previously experienced disease progression on or after fluoropyrimidine‐ or platinum‐containing chemotherapy.48

3.3. Antibody‐drug conjugate

Antibody‐drug conjugate (ADCs) is a kind of targeted drug that takes advantage of specifically binding properties of antibody and the high cytotoxicity of small molecule with the aim to improve the therapeutic efficacy of anti‐cancer drugs and minimize adverse effects.172, 173, 174 They often consist of one monoclonal antibody as recognition part, and a linker and chemical or cytotoxic agents as functional part.

Up to now, several ADCs have been investigated in breast cancer treatment. Among them, T‐DM1 is one of the most popular ADCs. In 2013, T‐DM1 received FDA approval for the treatment of HER2‐positive metastatic breast cancer or locally advanced breast cancer.31, 32, 175 T‐DM1 is composed of trastuzumab, a stable linker (MCC), and the cytotoxic agent derivative of maytansine, emtansine (DM1), which specifically binds to HER2.172, 173, 174, 175, 176, 177 It can reverse the trastuzumab resistance of breast cancer patients, showing great promising in the treatment of breast cancer patients who have failed on prior treatment with trastuzumab.32 T‐DM1 first transports emtansine to target cells via the recognition of monoclonal antibody trastuzumab to the HER2 protein of breast cancer cells. Then it is internalized by endocytosis and degraded into trastuzumab and DM1 by the lysosomal enzyme. Eventually, the DM1 is delivered and binds to tubulin, leading to the cell cycle arrest and apoptosis by destroying the microtubules. Meanwhile, trastuzumab can also perform its properties of ADCC and inhibit HER2‐mediated signalling pathways, providing a perfect double effect in breast cancer treatment.178, 179, 180 Several phase II studies of T‐DM1 monotherapy indicate that T‐DM1 will benefit metastatic breast cancer patients, who had failed at both HER2 targeted therapy and chemotherapy.181, 182, 183

3.4. Other targeted biopharmaceuticals for breast cancer treatment

With respect to the targeted biopharmaceuticals outlined above, other novel drugs such as denosumab,24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37 aflibercept,49, 50, 51 etc., also show great potential in breast cancer treatment.

3.4.1. Denosumab

Denosumab is the newest fully human monoclonal antibody of IgG2 subtype, which specifically binds to the receptor activator of nuclear factor kappa B ligand (RANKL).24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37 It can inhibit the receptor RANK on osteoclasts and block the downstream signalling pathways of breast cancer‐mediated bone disease.184, 185, 186, 187 It was first developed for osteoporosis in postmenopausal women at increased risk of fractures and bone loss induced by hormone ablation in prostate cancer patients. Afterwards, it was also used for the treatment of breast cancer patients with bone metastases.37 It had taken long time to conduct clinical trials of denosumab in breast cancer treatment before it got the FDA approval in 2010. Several randomized phase III trials of denosumab demonstrated that denosumab was non‐inferiority with improved tolerability, which showed great potential for the treatment of advanced breast cancer with bone metastases.26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 188, 189, 190

3.4.2. Aflibercept

Aflibercept is a recombinant peptide‐antibody fusion protein consisting of the extracellular domains of VEGF and Fc portion of immunoglobulin G1, which shows high affinity for all ligands that bind to these receptors, including VEGF‐A, VEGF‐B and placental growth factors.49, 50, 51, 191 The recent results demonstrated that aflibercept had got perfect anti‐tumour effects and anti‐angiogenic activity when used as a single agent or in combination therapy.52, 192, 193, 194 Two phase I dose‐escalation studies of aflibercept in patients with advanced solid tumours indicated that the dose recommended for further clinical development based on tolerability, anti‐tumour activity and pharmacokinetics was 6 mg/kg.192, 193 Notably, several phase II trials to evaluate aflibercept in patients with metastatic breast cancer treatment have been conducted.50, 195 Further experiments should be done to certificate its activity and efficacy before it is clinically used for breast cancer targeted therapy.

4. Challenges and strategies

In spite of significant progress towards breast cancer treatment made by targeted biopharmaceuticals in the past 20 years, breast cancer remains the second leading cause of death among women.2, 3, 4 Moreover, the side effects of biopharmaceuticals induced by the non‐specificity of biomarkers, which cannot efficiently distinguish cancer cells and normal cells, have often been reported in their clinic utilization.196 In addition, biopharmaceuticals could be degraded easily with a very short half‐life under physiological conditions, making the optimal therapeutical doses larger than that of other agents, which will hugely increase the cost of treatment.196, 197, 198 Moreover, the present biopharmacueticals are always heterogeneous products, which may cause serious immunological rejection response. The bioprocessing and purification technologies are another kind of obstacles that impede the development of bioparmaceuticals.199, 200

To circumvent those shortcomings of targeted biopharmaceuticals, all the therapeutical biopharmaceuticals should be humanized, redesigned or specifically modified, which are expected to be more tolerated and efficiently correlated with cancer targeted therapy. In order to improve the activity and affectivity of targeted biopharmaceuticals, more synergy therapies of biopharmaceuticals or in combination with cytotoxic agents should also be developed. In addition, developing optimal and innovative individual biotechnology for continuous bioprocessing and purification of biopharmaceuticals will greatly promote the industrialized production.201, 202 These studies will certainly overcome the challenges confronted by the clinic utilization of targeted biopharmaceuticals for breast cancer treatment.

5. Conclusion

Cell‐specific biomarkers are the foundation of breast cancer targeted therapy. During the past years, targeted biopharmaceuticals in breast cancer treatment has been demonstrated to possess obvious advantages over traditional therapeuticals due to their high affinity, specificity, selectivity, low toxicity as well as perfect efficacy. Since the first monoclonal antibody trastuzumab for the first‐line HER2‐positive metastatic breast cancer targeted therapy was approved by the FDA in 1998, there have been several targeted biopharmaceuticals developed for breast cancer treatment. Among all these novel biopharmaceuticals, some have been approved for clinical treatment such as trastuzumab, pertuzumab and T‐DM1, while others are still under clinical research. With respect to monotherapy, trials of targeted biopharmaceuticals in combination with cytotoxic agents are growing vigorously. In order to enhance treatment efficiency, it is very imperative to figure out the occurrence, development and prognosis of breast cancer, which will benefit the discovery of highly cell‐specific biomarkers of breast cancer.

Acknowledgements

This research is financially supported by the State Key Basic Research Program of the PRC (2014CB744501), the NSF of China (61271056, 61527806), Jiangsu Province Natural Science Foundation (BK20141332), the project funded by the Priority Academic Program Development of Jiangsu Higher Education Institutions, and the Fundamental Research Funds for the Central Universities.

References

  • 1. Colombo M, Corsi F, Foschi D, et al. HER2 targeting as a two‐sided strategy for breast cancer diagnosis and treatment: outlook and recent implications in nanomedical approaches. Pharmacol Res. 2010;62:150–165. [DOI] [PubMed] [Google Scholar]
  • 2. DeSantis CE, Fedewa SA, Goding SA, Kramer JL, Smith RA, Jemal A. Breast cancer statistics, 2015: convergence of incidence rates between black and white women. CA Cancer J Clin. 2016;66:31–42. [DOI] [PubMed] [Google Scholar]
  • 3. DeSantis CE, Siegel R, Bandi P, Jemal A. Breast cancer statistics, 2013. CA Cancer J Clin. 2014;64:52–62. [DOI] [PubMed] [Google Scholar]
  • 4. DeSantis CE, Siegel R, Bandi P, Jemal A. Breast cancer statistics, 2011. CA Cancer J Clin. 2011;61:408–418. [DOI] [PubMed] [Google Scholar]
  • 5. Hagberg KW, Taylor A, Hernandez RK, Jick S. Incidence of bone metastases in breast cancer patients in the United Kingdom: results of a multi‐database linkage study using the general practice research database. Cancer Epidemiol. 2013;37:240–246. [DOI] [PubMed] [Google Scholar]
  • 6. Hamdy AA, Amal SI. Breast cancer in Egypt, China and Chinese: statistics and beyond. J Thorac Dis. 2014;6:864–866. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Rosenblum D, Peer D. Omics‐based nanomedicine: the future of personalized oncology. Cancer Lett. 2014;352:126–136. [DOI] [PubMed] [Google Scholar]
  • 8. Lam SW, Jimenez CR, Boven E. Breast cancer classification by proteomic technologies: current state of knowledge. Cancer Treat Rev. 2014;40:129–138. [DOI] [PubMed] [Google Scholar]
  • 9. Ades F, Zardavas D, Bozovic‐Spasojevic I, et al. Luminal B breast cancer: molecular characterization, clinical management, and future perspectives. J Clin Oncol. 2014;32:2794–2803. [DOI] [PubMed] [Google Scholar]
  • 10. Yersal O, Barutca S. Biological subtypes of breast cancer: prognostic and therapeutic implications. World J Clin Oncol. 2014;5:412–424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Leidy J, Khan A, Kandil D. Basal‐like breast cancer update on clinicopathologic, immunohistochemical, and molecular features. Arch Pathol Lab Med. 2014;138:37–43. [DOI] [PubMed] [Google Scholar]
  • 12. Ahmed S, Sami A, Xiang J. HER2‐directed therapy: current treatment options for HER2‐positive breast cancer. Breast Cancer. 2015;22:101–116. [DOI] [PubMed] [Google Scholar]
  • 13. Kulhari H, Pooja D, Rompicharla SVK, Sistla R, Adams DJ. Biomedical applications of trastuzumab: as a therapeutic agent and a targeting ligand. Med Res Rev. 2015;35:849–876. [DOI] [PubMed] [Google Scholar]
  • 14. Figueroa‐Magalhaes MC, Jelovac D, Connolly RM, Wolff AC. Treatment of HER2‐positive breast cancer. Breast. 2014;23:128–136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Parkinson B, Pearson S, Viney R. Economic evaluations of trastuzumab in HER2‐positive metastatic breast cancer: a systematic review and critique. Eur J Health Econ. 2014;15:93–112. [DOI] [PubMed] [Google Scholar]
  • 16. Martin Martorell P, Bermejo De Las Heras B, Perez‐Fidalgo JA, et al. Subcutaneous trastuzumab: drug development and current position. Clin Transl Oncol. 2014;16:859–864. [DOI] [PubMed] [Google Scholar]
  • 17. Zhou L, Xu N, Sun Y, Liu XM. Targeted biopharmaceuticals for cancer treatment. Cancer Lett. 2014;352:145–151. [DOI] [PubMed] [Google Scholar]
  • 18. Kamada H. Proteomic analysis for the development of biopharmaceuticals. Yakugaku Zasshi. 2013;133:925–930. [DOI] [PubMed] [Google Scholar]
  • 19. Lingg N, Zhang P, Song Z, Bardor M. The sweet tooth of biopharmaceuticals: importance of recombinant protein glycosylation analysis. Biotechnol J. 2012;7:1462–1472. [DOI] [PubMed] [Google Scholar]
  • 20. Jang M, Yoon YI, Kwon YS, et al. Trastuzumab‐conjugated liposome‐coated fluorescent magnetic nanoparticles to target breast cancer. Korean J Radiol. 2014;15:411–422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Ross JS, Slodkowska EA, Symmans WF, Pusztai L, Ravdin PM, Hortobagyi GN. The HER‐2 receptor and breast cancer: ten years of targeted anti‐HER‐2 therapy and personalized medicine. Oncologist. 2009;14:320–368. [DOI] [PubMed] [Google Scholar]
  • 22. Murphy CG, Morris PG. Recent advances in novel targeted therapies for HER2‐positive breast cancer. Anticancer Drugs. 2012;23:765–776. [DOI] [PubMed] [Google Scholar]
  • 23. Ceran C, Cokol M, Cingoz S, Tasan I, Ozturk M, Yagci T. Novel anti‐HER2 monoclonal antibodies: synergy and antagonism with tumor necrosis factor‐alpha. BMC Cancer. 2012;12:450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Fleeman N, Bagust A, Beale S, et al. Pertuzumab in combination with trastuzumab and docetaxel for the treatment of HER2‐positive metastatic or locally recurrent unresectable breast cancer. Pharmacoeconomics. 2015;33:13–23. [DOI] [PubMed] [Google Scholar]
  • 25. Hudis CA. Drug therapy: trastuzumab‐mechanism of action and use in clinical practice. N Engl J Med. 2007;357:39–51. [DOI] [PubMed] [Google Scholar]
  • 26. Singh JC, Jhaveri K, Esteva FJ. HER2‐positive advanced breast cancer: optimizing patient outcomes and opportunities for drug development. Br J Cancer. 2014;111:1888–1898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Kawajiri H, Takashima T, Kashiwagi S, Noda S, Onoda N, Hirakawa K. Pertuzumab in combination with trastuzumab and docetaxel for HER2‐positive metastatic breast cancer. Expert Rev Anticancer Ther. 2015;15:17–26. [DOI] [PubMed] [Google Scholar]
  • 28. Malenfant SJ, Eckmann KR, Barnett CM. Pertuzumab: a new targeted therapy for HER2‐positive metastatic breast cancer. Pharmacotherapy. 2014;34:60–71. [DOI] [PubMed] [Google Scholar]
  • 29. Swain SM, Baselga J, Kim S, et al. Pertuzumab, trastuzumab, and docetaxel in HER2‐positive metastatic breast cancer. N Engl J Med. 2015;372:724–734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Leung W, Roxanis I, Sheldon H, et al. Combining lapatinib and pertuzumab to overcome lapatinib resistance due to NRG1‐mediated signalling in HER2‐amplified breast cancer. Oncotarget. 2015;6:5678–5694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Baron JM, Boster BL, Barnett CM. Ado‐trastuzumab emtansine (T‐DM1): a novel antibody‐drug conjugate for the treatment of HER2‐positive metastatic breast cancer. J Oncol Pharm Pract. 2015;21:132–142. [DOI] [PubMed] [Google Scholar]
  • 32. Van den Mooter T, Teuwen L, Rutten A, Dirix L. Trastuzumab emtansine in advanced human epidermal growth factor receptor 2‐positive breast cancer. Expert Opin Biol Ther. 2015;15:749–760. [DOI] [PubMed] [Google Scholar]
  • 33. Girish S, Gupta M, Wang B, et al. Clinical pharmacology of trastuzumab emtansine (T‐DM1): an antibody‐drug conjugate in development for the treatment of HER2‐positive cancer. Cancer Chemother Pharmacol. 2012;69:1229–1240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Boyraz B, Sendur MAN, Aksoy S, et al. Trastuzumab emtansine (T‐DM1) for HER2‐positive breast cancer. Curr Med Res Opin. 2013;29:405–414. [DOI] [PubMed] [Google Scholar]
  • 35. Rimawi MF, Schiff R, Osborne CF. Targeting HER2 for the treatment of breast cancer. Annu Rev Med. 2015;66:111–128. [DOI] [PubMed] [Google Scholar]
  • 36. Lee BL, Higgins MJ, Goss PE. Denosumab and the current status of bone‐modifying drugs in breast cancer. Acta Oncol. 2012;51:157–167. [DOI] [PubMed] [Google Scholar]
  • 37. Casas A, Llombart A, Martin M. Denosumab for the treatment of bone metastases in advanced breast cancer. Breast. 2013;22:585–592. [DOI] [PubMed] [Google Scholar]
  • 38. Galluzzi L, Vacchelli E, Fridman WH, et al. Trial watch monoclonal antibodies in cancer therapy. Oncoimmunology. 2012;1:28–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Haense N, Jaeger M, Klunker D, et al. Phase I/II, open label, dose escalating study to investigate safety, tolerability, and preliminary efficacy of the trifunctional anti‐HER2/neu x anti‐CD3 antibody ertumaxomab in patients with HER2/neu (1+/SISH positive, 2+and 3+) expressing solid tumors progressing after standard therapy. Oncol Res Treat. 2014;371:106. [Google Scholar]
  • 40. Diermeier‐Daucher S, Ortmann O, Buchholz S, Brockhoff G. Trifunctional antibody ertumaxomab non‐immunological effects on HER2 receptor activity and downstream signaling. MAbs. 2012;4:614–622. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Kiewe P, Thiel E. Ertumaxomab: a trifunctional antibody for breast cancer treatment. Expert Opin Investig Drugs. 2008;17:1553–1558. [DOI] [PubMed] [Google Scholar]
  • 42. Ranieri G, Patruno R, Ruggieri E, Montemurro S, Valerio P, Ribatti D. Vascular endothelial growth factor (VEGF) as a target of bevacizumab in cancer: from the biology to the clinic. Curr Med Chem. 2006;13:1845–1857. [DOI] [PubMed] [Google Scholar]
  • 43. Kumler I, Christiansen OG, Nielsen DL. A systematic review of bevacizumab efficacy in breast cancer. Cancer Treat Rev. 2014;40:960–979. [DOI] [PubMed] [Google Scholar]
  • 44. Gropper AB, Mayer EL. Anti‐angiogenic strategies in breast cancer: an update. Curr Breast Cancer Rep. 2010;2:174–181. [Google Scholar]
  • 45. Manso L, Moreno F, Marquez R, et al. Use of bevacizumab as a first‐line treatment for metastatic breast cancer. Curr Oncol. 2015;22:E51–E60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Miller KD, Chap LI, Holmes FA, et al. Randomized phase III trial of capecitabine compared with bevacizumab plus capecitabine in patients with previously treated metastatic breast cancer. J Clin Oncol. 2005;23:792–799. [DOI] [PubMed] [Google Scholar]
  • 47. Aprile G, Fontanella C, Rihawi K, Grossi F, Rijavec E. Ramucirumab: preclinical research and clinical development. Onco Targets Ther. 2014;7:1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Poole RM, Vaidya A. Ramucirumab: first global approval. Drugs. 2014;74:1047–1058. [DOI] [PubMed] [Google Scholar]
  • 49. Schmid MK, Bachmann LM, Fas L, Kessels AG, Job OM, Thiel MA. Efficacy and adverse events of aflibercept, ranibizumab and bevacizumab in age‐related macular degeneration: a trade‐off analysis. Br J Ophthalmol. 2015;99:141–146. [DOI] [PubMed] [Google Scholar]
  • 50. Perez EA, Spano J. Current and emerging targeted therapies for metastatic breast cancer. Cancer. 2012;118:3014–3025. [DOI] [PubMed] [Google Scholar]
  • 51. Le Saux O, You B, Freyer G. Antiangiogenic therapy in patients with HER2‐positive metastatic breast cancer: a case series. Clin Breast Cancer. 2014;14:e89–e94. [DOI] [PubMed] [Google Scholar]
  • 52. Lockhart AC, Rothenberg ML, Dupont J, et al. Phase I study of intravenous vascular endothelial growth factor trap, aflibercept, in patients with advanced solid tumors. J Clin Oncol. 2010;28:207–214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Toss A, Cristofanilli M. Molecular characterization and targeted therapeutic approaches in breast cancer. Breast Cancer Res. 2015;17:60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Lordick F, Schulze T, Jäger D. The role of biologics in stomach cancer. Target Oncol. 2008;3:71–79. [Google Scholar]
  • 55. Zhao X, Rodland EA, Tibshirani R, Plevritis S. Molecular subtyping for clinically defined breast cancer subgroups. Breast Cancer Res. 2015;17:29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Lai X, Chen S. Identification of novel biomarker candidates for immunohistochemical diagnosis to distinguish low‐grade chondrosarcoma from enchondroma. Proteomics. 2015;15:2358–2368. [DOI] [PubMed] [Google Scholar]
  • 57. Jiang J, Liu H, Liu Z, Tan S, Wu B. Identification of cystatin SN as a novel biomarker for pancreatic cancer. Tumor Biol. 2015;36:3903–3910. [DOI] [PubMed] [Google Scholar]
  • 58. Yoneyama K, Kojima S, Kodani Y, et al. Proteomic identification of autoantibodies in sera from patients with ovarian cancer as possible diagnostic biomarkers. Anticancer Res. 2015;35:881–889. [PubMed] [Google Scholar]
  • 59. Lee MJ, Na K, Jeong S, et al. Identification of human complement factor b as a novel biomarker candidate for pancreatic ductal adenocarcinoma. J Proteome Res. 2014;13:4878–4888. [DOI] [PubMed] [Google Scholar]
  • 60. Kim H, Woo SJ, Suh EJ, et al. Identification of vinculin as a potential plasma marker for age‐related macular degeneration. Invest Ophthalmol Vis Sci. 2014;55:7166–7176. [DOI] [PubMed] [Google Scholar]
  • 61. Li Y, Zhang Q, Peng B, Shao Q, Qian W, Zhang J. Identification of glutathione S‐transferase omega 1 (GSTO1) protein as a novel tumor‐associated antigen and its autoantibody in human esophageal squamous cell carcinoma. Tumor Biol. 2014;35:10871–10877. [DOI] [PubMed] [Google Scholar]
  • 62. Cheng Y, Chen Y, Sun X, et al. Identification of potential serum biomarkers for rheumatoid arthritis by high‐resolution quantitative proteomic analysis. Inflammation. 2014;37:1459–1467. [DOI] [PubMed] [Google Scholar]
  • 63. Banerji S, Cibulskis K, Rangel‐Escareno C, Brown KK, Carter SL, Frederick AM. Sequence analysis of mutations and translocations across breast cancer subtypes. Nature. 2012;486:405–409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Voduc KD, Cheang MCU, Tyldesley S, Gelmon K, Nielsen TO, Kennecke H. Breast cancer subtypes and the risk of local and regional relapse. J Clin Oncol. 2010;28:1684–1691. [DOI] [PubMed] [Google Scholar]
  • 65. Maleki S, Dorokhova O, Sunkara J, Schlesinger K, Suhrland M, Oktay MH. Estrogen, progesterone, and HER‐2 receptor immunostaining in cytology. Diagn Cytopathol. 2013;41:864–870. [DOI] [PubMed] [Google Scholar]
  • 66. Curtis C, Shah SP, Chin S, Turashvili G, Rueda OM, Dunning MJ. The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature. 2012;486:346–352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Osborne CK. Steroid hormone receptors in breast cancer management. Breast Cancer Res Treat. 1998;51:227–238. [DOI] [PubMed] [Google Scholar]
  • 68. Thakkar JP, Mehta DG. A review of an unfavorable subset of breast cancer: estrogen receptor positive progesterone receptor negative. Oncologist. 2011;16:276–285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Osborne CK, Shou J, Massarweh S, Schiff R. Crosstalk between estrogen receptor and growth factor receptor pathways as a cause for endocrine therapy resistance in breast cancer. Clin Cancer Res. 2005;11:865S–870S. [PubMed] [Google Scholar]
  • 70. Dunbier AK, Anderson H, Folkerd E, et al. Expression of estrogen responsive genes in breast cancers correlates with plasma estradiol levels in postmenopausal women. Cancer Res. 2009;69:78S. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Cui X, Schiff R, Arpino G, Osborne CK, Lee AV. Biology of progesterone receptor loss in breast cancer and its implications for endocrine therapy. J Clin Oncol. 2005;23:7721–7735. [DOI] [PubMed] [Google Scholar]
  • 72. Gschwind A, Fischer OM, Ullrich A. Timeline‐the discovery of receptor tyrosine kinases: targets for cancer therapy. Nat Rev Cancer. 2004;4:361–370. [DOI] [PubMed] [Google Scholar]
  • 73. Hurvitz SA, Hu Y, O Brien N, Finn RS. Current approaches and future directions in the treatment of HER2‐positive breast cancer. Cancer Treat Rev. 2013;39:219–229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Ross JS, Fletcher JA, Bloom KJ, et al. Targeted therapy in breast cancer – the HER‐2/neu gene and protein. Mol Cell Proteomics. 2004;3:379–398. [DOI] [PubMed] [Google Scholar]
  • 75. Ross JS, Fletcher JA, Linette GP, Stec J, Clark E, Ayers M. The HER‐2/neu gene and protein in breast cancer 2003: biomarker and target of therapy. Oncologist. 2003;8:307–325. [DOI] [PubMed] [Google Scholar]
  • 76. Cho HS, Mason K, Ramyar KX, et al. Structure of the extracellular region of HER2 alone and in complex with the Herceptin Fab. Nature. 2003;421:756–760. [DOI] [PubMed] [Google Scholar]
  • 77. Veronesi U, Zurrida S, Goldhirsch A, Rotmensz N, Viale G. Breast cancer classification: time for a change. J Clin Oncol. 2009;27:2427–2428. [DOI] [PubMed] [Google Scholar]
  • 78. van Ramshorst MS, Sonke GS. Treatment of HER2‐positive metastatic breast cancer. N Engl J Med. 2015;372:1964. [DOI] [PubMed] [Google Scholar]
  • 79. Fan X, Brezski RJ, Fa M, et al. A single proteolytic cleavage within the lower hinge of trastuzumab reduces immune effector function and in vivo efficacy. Breast Cancer Res. 2012;14:R116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Li J, Chen F, Cona MM, et al. A review on various targeted anticancer therapies. Target Oncol. 2012;7:69–85. [DOI] [PubMed] [Google Scholar]
  • 81. Bose R, Kavuri SM, Searleman AC, et al. Activating HER2 mutations in HER2 gene amplification negative breast cancer. Cancer Discov. 2013;3:224–237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Wolff AC, Hammond MEH, Hicks DG, et al. Recommendations for human epidermal growth factor receptor 2 testing in breast cancer American society of clinical oncology/college of American pathologists clinical practice guideline update. Arch Pathol Lab Med. 2014;138:241–256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Banerjee S, Dowsett M, Ashworth A, Martin L. Mechanisms of disease: angiogenesis and the management of breast cancer. Nat Clin Pract Oncol. 2007;4:536–550. [DOI] [PubMed] [Google Scholar]
  • 84. Claesson‐Welsh L, Welsh M. VEGFA and tumour angiogenesis. J Intern Med. 2013;273:114–127. [DOI] [PubMed] [Google Scholar]
  • 85. Ferrara N, Gerber HP, LeCouter J. The biology of VEGF and its receptors. Nat Med. 2003;9:669–676. [DOI] [PubMed] [Google Scholar]
  • 86. Bozza C, Fontanella C, Buoro V, Mansutti M, Aprile G. Novel antiangiogenic drugs for the management of breast cancer: new approaches for an old issue? Expert Rev Clin Pharmacol. 2015;8:251–265. [DOI] [PubMed] [Google Scholar]
  • 87. Takahashi T, Yamaguchi S, Chida K, Shibuya M. A single autophosphorylation site on KDR/Flk‐1 is essential for VEGF‐A‐dependent activation of PLC‐gamma and DNA synthesis in vascular endothelial cells. EMBO J. 2001;20:2768–2778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Kerbel RS. Tumor angiogenesis – reply. N Engl J Med. 2008;359:764. [DOI] [PubMed] [Google Scholar]
  • 89. van der Meel R, Symons MH, Kudernatsch R, et al. The VEGF/Rho GTPase signalling pathway: a promising target for anti‐angiogenic/anti‐invasion therapy. Drug Discov Today. 2011;16:219–228. [DOI] [PubMed] [Google Scholar]
  • 90. Fantozzi A, Gruber DC, Pisarsky L, et al. VEGF‐mediated angiogenesis links EMT‐induced cancer stemness to tumor initiation. Cancer Res. 2013;74:1566–1575. [DOI] [PubMed] [Google Scholar]
  • 91. Goel HL, Mercurio AM. VEGF targets the tumour cell. Nat Rev Cancer. 2013;13:871–882. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Miller K, Wang M, Gralow J, et al. Paclitaxel plus bevacizumab versus paclitaxel alone for metastatic breast cancer. N Engl J Med. 2007;357:2666–2676. [DOI] [PubMed] [Google Scholar]
  • 93. Wadhwa R, Elimova E, Shiozaki H, Sudo K, Blum MA, Estrella JS. Anti‐angiogenic agent ramucirumab: meaningful or marginal? Expert Rev Anticancer. 2014;14:367–379. [DOI] [PubMed] [Google Scholar]
  • 94. Sharma T, Dhingra R, Singh S, et al. Aflibercept: a novel VEGF targeted agent to explore the future perspectives of anti‐angiogenic therapy for the treatment of multiple tumors. Mini Rev Med Chem. 2013;13:530–540. [DOI] [PubMed] [Google Scholar]
  • 95. Campbell MR, Moasser MM. HER targeting in HER2‐negative breast cancers: looking for the HER3 positive. Clin Cancer Res. 2015;21:2886–2888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Raina D, Uchida Y, Kharbanda A, et al. Targeting the MUC1‐C oncoprotein downregulates HER2 activation and abrogates trastuzumab resistance in breast cancer cells. Oncogene. 2014;33:3422–3431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Rota LM, Wood TL. Crosstalk of the insulin‐like growth factor receptor with the Wnt signaling pathway in breast cancer. Front Endocrinol. 2015;6:92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Shak S. Overview of the trastuzumab (herceptin) anti‐HER2 monoclonal antibody clinical program in HER2‐overexpressing metastatic breast cancer. Semin Oncol. 1999;26:71–77. [PubMed] [Google Scholar]
  • 99. Kurebayashi J, Kanomata N, Yamashita T, et al. Prognostic value of phosphorylated HER2 in HER2‐positive breast cancer patients treated with adjuvant trastuzumab. Breast Cancer. 2015;22:292–299. [DOI] [PubMed] [Google Scholar]
  • 100. Vogel CL, Cobleigh MA, Tripathy D, et al. Efficacy and safety of trastuzumab as a single agent in first‐line treatment of HER2‐overexpressing metastatic breast cancer. J Clin Oncol. 2002;20:719–726. [DOI] [PubMed] [Google Scholar]
  • 101. Nahta R, Esteva FJ. Herceptin: mechanisms of action and resistance. Cancer Lett. 2006;232:123–138. [DOI] [PubMed] [Google Scholar]
  • 102. Nagata Y, Lan KH, Zhou XY, et al. PTEN activation contributes to tumor inhibition by trastuzumab, and loss of PTEN predicts trastuzumab resistance in patients. Cancer Cell. 2004;6:117–127. [DOI] [PubMed] [Google Scholar]
  • 103. Arnould L, Gelly M, Penault‐Llorca F, et al. Trastuzumab‐based treatment of HER2‐positive breast cancer: an antibody‐dependent cellular cytotoxicity mechanism? Br J Cancer. 2006;94:259–267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Albanell J, Codony J, Rovira A, Mellado B, Gascon P. Mechanism of action of anti‐HER2 monoclonal antibodies: scientific update on trastuzumab and 2C4. Adv Exp Med Biol. 2003;532:253–268. [DOI] [PubMed] [Google Scholar]
  • 105. Izumi Y, Xu L, di Tomaso E, Fukumura D, Jain RK. Tumor biology‐herceptin acts as an anti‐angiogenic cocktail. Nature. 2002;416:279–280. [DOI] [PubMed] [Google Scholar]
  • 106. Pegram CHYA. Anti‐erbB‐2 antibody trastuzumab in the treatment of HER2‐amplified breast cancer. Invest New Drugs. 2005;23:391–409. [DOI] [PubMed] [Google Scholar]
  • 107. Slamon DJ, Leyland‐Jones B, Shak S, et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med. 2001;344:783–792. [DOI] [PubMed] [Google Scholar]
  • 108. Soularue E, Cohen R, Tournigand C, et al. Efficacy and safety of trastuzumab in combination with oxaliplatin and fluorouracil‐based chemotherapy for patients with HER2‐positive metastatic gastric and gastro‐oesophageal junction adenocarcinoma patients: a retrospective study. Bull Cancer. 2015;102:324–331. [DOI] [PubMed] [Google Scholar]
  • 109. Haag G, Apostolidis L, Jaeger D. Efficacy and safety of trastuzumab‐based therapy in combination with different chemotherapeutic regimens in advanced esophagogastric cancer‐a single cancer‐center experience. Tumori. 2014;100:237–242. [DOI] [PubMed] [Google Scholar]
  • 110. Anonymous . Abstracts of the 10th international congress of the metastasis research society, Genoa, Italy, 17–20 September 2004. Clin Exp Metastas. 2004;21:599–662. [Google Scholar]
  • 111. Nahta R, Hung MC, Esteva FJ. The HER‐2‐targeting antibodies trastuzumab and pertuzumab synergistically inhibit the survival of breast cancer cells. Cancer Res. 2004;64:2343–2346. [DOI] [PubMed] [Google Scholar]
  • 112. Nagy P, Friedlander E, Tanner M, et al. Decreased accessibility and lack of activation of ErbB2 in JIMT‐1, a herceptin‐resistant, MUC4‐expressing breast cancer cell line. Cancer Res. 2005;65:473–482. [PubMed] [Google Scholar]
  • 113. Nielsen DL, Andersson M, Kamby C, et al. HER2‐targeted therapy in breast cancer. Monoclonal antibodies and tyrosine kinase inhibitors. Cancer Treat Rev. 2009;35:121–136. [DOI] [PubMed] [Google Scholar]
  • 114. Koukourakis MI, Giatromanolaki A, Bottini A, et al. Prospective neoadjuvant analysis of PET imaging and mechanisms of resistance to Trastuzumab shows role of HIF1 and autophagy. Br J Cancer. 2014;110:2209–2216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Thery J, Spano J, Azria D, Raymond E, Llorca FP. Resistance to human epidermal growth factor receptor type 2‐targeted therapies. Eur J Cancer. 2014;50:892–901. [DOI] [PubMed] [Google Scholar]
  • 116. Calaf GM, Zepeda AB, Castillo RL, et al. Molecular aspects of breast cancer resistance to drugs. Int J Oncol. 2015;47:437–445. [DOI] [PubMed] [Google Scholar]
  • 117. Yamaguchi H, Chang S, Hsu JL, Hung M. Signaling cross‐talk in the resistance to HER family receptor targeted therapy. Oncogene. 2014;33:1073–1081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Perez EA, Romond EH, Suman VJ, et al. Four‐year follow‐up of trastuzumab plus adjuvant chemotherapy for operable human epidermal growth factor receptor 2‐positive breast cancer: joint analysis of data from NCCTG N9831 and NSABP B‐31. J Clin Oncol. 2011;29:3366–3373. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. McKeage K, Perry CM, et al. Trastuzumab‐a review of its use in the treatment of metastatic breast cancer overexpressing HER2. Drugs. 2002;62:209–243. [DOI] [PubMed] [Google Scholar]
  • 120. Cobleigh MA, Vogel CL, Tripathy D, et al. Multinational study of the efficacy and safety of humanized anti‐HER2 monoclonal antibody in women who have HER2‐overexpressing metastatic breast cancer that has progressed after chemotherapy for metastatic disease. J Clin Oncol. 1999;17:2639–2648. [DOI] [PubMed] [Google Scholar]
  • 121. Lei N, Gong C, Qian Z, et al. Therapeutic application of injectable thermosensitive hydrogel in preventing local breast cancer recurrence and improving incision wound healing in a mouse model. Nanoscale. 2012;4:5686–5693. [DOI] [PubMed] [Google Scholar]
  • 122. Baselga J, Cortes J, Kim S, et al. Pertuzumab plus trastuzumab plus docetaxel for metastatic breast cancer. N Engl J Med. 2012;366:109–119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Quartino AL, Li H, Jin JY, et al. Pharmacokinetic and exposure‐response analyses of pertuzumab plus trastuzumab and docetaxel during neoadjuvant treatment of HER2+ early breast cancer. Clin Pharmacol Ther. 2014;951:S6–S7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Camidge DR, Berge EM, Doebele RC, et al. A phase ii, open‐label study of ramucirumab in combination with paclitaxel and carboplatin as first‐line therapy in patients with stage iiib/iv non‐small‐cell lung cancer. J Thorac Oncol. 2014;9:1532–1539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. Lin NU, Eierman W, Greil R, et al. Randomized phase II study of lapatinib plus capecitabine or lapatinib plus topotecan for patients with HER2‐positive breast cancer brain metastases. J Neurooncol. 2011;105:613–620. [DOI] [PubMed] [Google Scholar]
  • 126. Welslau M, Dieras V, Sohn J, et al. Patient‐reported outcomes from emilia, a randomized phase 3 study of trastuzumab emtansine (T‐DM1) versus capecitabine and lapatinib in human epidermal growth factor receptor 2‐positive locally advanced or metastatic breast cancer. Cancer. 2014;120:642–651. [DOI] [PubMed] [Google Scholar]
  • 127. Hu X, Zhang J, Xu B, et al. Cisplatin plus gemcitabine versus paclitaxel plus gemcitabine as first‐line therapy for metastatic triple‐negative breast cancer (CBCSG006): a randomised, open‐label, multicentre, phase 3 trial. Lancet Oncol. 2015;16:436–446. [DOI] [PubMed] [Google Scholar]
  • 128. Salvador J, Manso L, de la Haba J, et al. Final results of a phase II study of paclitaxel, bevacizumab, and gemcitabine as first‐line therapy for patients with HER2‐negative metastatic breast cancer. Clin Transl Oncol. 2015;17:160–166. [DOI] [PubMed] [Google Scholar]
  • 129. Silver DP, Richardson AL, Eklund AC, et al. Efficacy of neoadjuvant cisplatin in triple‐negative breast cancer. J Clin Oncol. 2010;28:1145–1153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Franklin MC, Carey KD, Vajdos FF, Leahy DJ, de Vos AM, Sliwkowski MX. Insights into ErbB signaling from the structure of the ErbB2‐pertuzumab complex. Cancer Cell. 2004;5:317–328. [DOI] [PubMed] [Google Scholar]
  • 131. Agus DB, Gordon MS, Taylor C, et al. Phase I clinical study of pertuzumab, a novel HER dimerization inhibitor, in patients with advanced cancer. J Clin Oncol. 2005;23:2534–2543. [DOI] [PubMed] [Google Scholar]
  • 132. Chung C, Lam MSH. Pertuzumab for the treatment of human epidermal growth factor receptor type 2‐positive metastatic breast cancer. Am J Health Syst Pharm. 2013;70:1579–1587. [DOI] [PubMed] [Google Scholar]
  • 133. Agus DB, Akita RW, Fox WD, et al. Targeting ligand‐activated ErbB2 signaling inhibits breast and prostate tumor growth. Cancer Cell. 2002;2:127–137. [DOI] [PubMed] [Google Scholar]
  • 134. Park S, Jiang Z, Mortenson ED, et al. The therapeutic effect of anti‐her2/neu antibody depends on both innate and adaptive immunity. Cancer Cell. 2010;18:160–170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Yarden Y, Sliwkowski MX. Untangling the ErbB signalling network. Nat Rev Mol Cell Biol. 2001;2:127–137. [DOI] [PubMed] [Google Scholar]
  • 136. De Mattos‐Arruda L, Cortes J. Use of pertuzumab for the treatment of HER2‐positive metastatic breast cancer. Adv Ther. 2013;30:645–658. [DOI] [PubMed] [Google Scholar]
  • 137. Scheuer W, Friess T, Burtscher H, Bossenmaier B, Endl J, Hasmann M. Strongly enhanced antitumor activity of trastuzumab and pertuzumab combination treatment on HER2‐positive human xenograft tumor models. Cancer Res. 2009;69:9330–9336. [DOI] [PubMed] [Google Scholar]
  • 138. Cortes J, Fumoleau P, Bianchi GV, et al. Pertuzumab monotherapy after trastuzumab‐based treatment and subsequent reintroduction of trastuzumab: activity and tolerability in patients with advanced human epidermal growth factor receptor 2‐positive breast cancer. J Clin Oncol. 2012;30:1594–1600. [DOI] [PubMed] [Google Scholar]
  • 139. Baselga J, Gelmon KA, Verma S, et al. Phase II trial of pertuzumab and trastuzumab in patients with human epidermal growth factor receptor 2‐positive metastatic breast cancer that progressed during prior trastuzumab therapy. J Clin Oncol. 2010;28:1138–1144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Bodmer M, Meier C, Krahenbuehl S, Jick SS, Meier CR. Long‐term metformin use is associated with decreased risk of breast cancer. Diabetes Care. 2010;33:1304–1308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Ayres LR, de Almeida Campos MS, de Oliveira Gozzo T, et al. Trastuzumab induced cardiotoxicity in HER2 positive breast cancer patients attended in a tertiary hospital. Int J Clin Pharm. 2015;37:365–372. [DOI] [PubMed] [Google Scholar]
  • 142. Gianni L, Pienkowski T, Im Y, et al. Efficacy and safety of neoadjuvant pertuzumab and trastuzumab in women with locally advanced, inflammatory, or early HER2‐positive breast cancer (NeoSphere): a randomised multicentre, open‐label, phase 2 trial. Lancet Oncol. 2012;13:25–32. [DOI] [PubMed] [Google Scholar]
  • 143. Arrondeau J, Cottu P. Pertuzumab plus trastuzumab plus docetaxel for metastatic breast cancer. Oncologie. 2012;14:351–352. [Google Scholar]
  • 144. Swain SM, Ewer MS, Cortes J, et al. Cardiac tolerability of pertuzumab plus trastuzumab plus docetaxel in patients with HER2‐positive metastatic breast cancer in cleopatra: a randomized, double‐blind, placebo‐controlled phase iii study. Oncologist. 2013;18:257–264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. Kiewe P, Hasmueller S, Kahlert S, et al. Phase I trial of the trifunctional anti‐HER2 x anti‐CD3 antibody ertumaxomab in metastatic breast cancer. Clin Cancer Res. 2006;12:3085–3091. [DOI] [PubMed] [Google Scholar]
  • 146. Zeidler R, Mysliwietz J, Csanady M, et al. The Fc‐region of a new class of intact bispecific antibody mediates activation of accessory cells and NK cells and induces direct phagocytosis of tumour cells. Br J Cancer. 2000;83:261–266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. Ruf P, Lindhofer H. Induction of a long‐lasting antitumor immunity by a trifunctional bispecific antibody. Blood. 2001;98:2526–2534. [DOI] [PubMed] [Google Scholar]
  • 148. Haense N, Pauligk C, Marme F, et al. Interim analysis of a phase I/II open label, dose escalating study to investigate safety, tolerability, and preliminary efficacy of the trifunctional anti‐HER2/neu x anti‐CD3 antibody ertumaxomab in patients with HER2/neu expressing solid tumors progressing after standard therapy. Oncol Res Treat. 2014;375:311–312. [Google Scholar]
  • 149. Cannita K, Paradisi S, Cocciolone V, et al. Bevacizumab/paclitaxel as first line therapy for metastatic breast cancer: new schedule in real life. Breast. 2015;241:S41. [Google Scholar]
  • 150. Li J, Yao Y, Jing J. Comment on Wang et al.: the efficacy of bevacizumab plus paclitaxel as first‐line treatment for HER2‐negative metastatic breast cancer: a meta‐analysis of randomized controlled trials. Tumor Biol. 2014;35:6183. [DOI] [PubMed] [Google Scholar]
  • 151. Kristensen T, Knutsson M, Wehland M, et al. Anti‐vascular endothelial growth factor therapy in breast cancer. Int J Mol Sci. 2014;15:23024–23041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152. Choueiri TK, Mayer EL, Je Y, et al. Congestive heart failure risk in patients with breast cancer treated with bevacizumab. J Clin Oncol. 2011;29:632–638. [DOI] [PubMed] [Google Scholar]
  • 153. Fralick M, Ray M, Fung C, Booth CM, Mallick R, Clemons MJ. Bevacizumab for advanced breast cancer: hope, hype, and hundreds of headlines. Oncologist. 2013;18:1174–1179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154. Dirix LY, Reynolds AR. Bevacizumab beyond progression in breast cancer. Lancet Oncol. 2014;15:1190–1191. [DOI] [PubMed] [Google Scholar]
  • 155. Wehland M, Bauer J, Infanger M, Grimm D. Target‐based anti‐angiogenic therapy in breast cancer. Curr Pharm Design. 2012;18:4244–4257. [DOI] [PubMed] [Google Scholar]
  • 156. von Minckwitz G, Loibl S, Untch M, et al. Survival after neoadjuvant chemotherapy with or without bevacizumab or everolimus for HER2‐negative primary breast cancer (GBG 44‐GeparQuinto). Ann Oncol. 2014;25:2363–2372. [DOI] [PubMed] [Google Scholar]
  • 157. von Minckwitz G, Eidtmann H, Rezai M, et al. Neoadjuvant chemotherapy and bevacizumab for HER2‐negative breast cancer. N Engl J Med. 2012;366:299–309. [DOI] [PubMed] [Google Scholar]
  • 158. Kontopodis E, Kentepozidis N, Christophyllakis C, et al. Docetaxel, gemcitabine and bevacizumab as salvage chemotherapy for HER‐2‐negative metastatic breast cancer. Cancer Chemother Pharmacol. 2015;75:153–160. [DOI] [PubMed] [Google Scholar]
  • 159. Yardley DA, Hart L, Waterhouse D, et al. Addition of bevacizumab to three docetaxel regimens as adjuvant therapy for early stage breast cancer. Breast Cancer Res Treat. 2013;142:655–665. [DOI] [PubMed] [Google Scholar]
  • 160. Curigliano G, Bagnardi V, Bertolini F, et al. Antiangiogenic therapy in recurrent breast cancer with lymphangitic spread to the chest wall: a randomized phase II trial of bevacizumab with sequential or concurrent oral vinorelbine and capecitabine. Breast. 2015;24:263–271. [DOI] [PubMed] [Google Scholar]
  • 161. Fang Y, Qu X, Cheng B, et al. The efficacy and safety of bevacizumab combined with chemotherapy in treatment of HER2‐negative metastatic breast cancer: a meta‐analysis based on published phase III trials. Tumor Biol. 2015;36:1933–1941. [DOI] [PubMed] [Google Scholar]
  • 162. Gligorov J, Doval D, Bines J, et al. Maintenance capecitabine and bevacizumab versus bevacizumab alone after initial first‐line bevacizumab and docetaxel for patients with HER2‐negative metastatic breast cancer (IMELDA): a randomised, open‐label, phase 3 trial. Lancet Oncol. 2014;15:1351–1360. [DOI] [PubMed] [Google Scholar]
  • 163. Pierga J, Delva R, Pivot X, et al. Bevacizumab and taxanes in the first‐line treatment of metastatic breast cancer: overall survival and subgroup analyses of the ATHENA study in France. Bull Cancer. 2014;101:780–788. [DOI] [PubMed] [Google Scholar]
  • 164. Dieras V, Wildiers H, Jassem J, et al. Trebananib (AMG 386) plus weekly paclitaxel with or without bevacizumab as first‐line therapy for HER2‐negative locally recurrent or metastatic breast cancer: a phase 2 randomized study. Breast. 2015;24:182–190. [DOI] [PubMed] [Google Scholar]
  • 165. Robert NJ, Dieras V, Glaspy J, et al. RIBBON‐1: randomized, double‐blind, placebo‐controlled, phase III trial of chemotherapy with or without bevacizumab for first‐line treatment of human epidermal growth factor receptor 2‐negative, locally recurrent or metastatic breast cancer. J Clin Oncol. 2011;29:1252–1260. [DOI] [PubMed] [Google Scholar]
  • 166. Brufsky AM, Hurvitz S, Perez E, et al. RIBBON‐2: a randomized, double‐blind, placebo‐controlled, phase III trial evaluating the efficacy and safety of bevacizumab in combination with chemotherapy for second‐line treatment of human epidermal growth factor receptor 2‐negative metastatic breast cancer. J Clin Oncol. 2011;29:4286–4293. [DOI] [PubMed] [Google Scholar]
  • 167. Miles DW, Dieras V, Cortes J, Duenne AA, Yi J, O'Shaughnessy J. First‐line bevacizumab in combination with chemotherapy for HER2‐negative metastatic breast cancer: pooled and subgroup analyses of data from 2447 patients. Ann Oncol. 2013;24:2773–2780. [DOI] [PubMed] [Google Scholar]
  • 168. Brodowicz T, Lang I, Kahan Z, et al. Selecting first‐line bevacizumab‐containing therapy for advanced breast cancer: turandot risk factor analyses. Br J Cancer. 2014;111:2051–2057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169. von Minckwitz G, Puglisi F, Cortes J, et al. Bevacizumab plus chemotherapy versus chemotherapy alone as second‐line treatment for patients with HER2‐negative locally recurrent or metastatic breast cancer after first‐line treatment with bevacizumab plus chemotherapy (TANIA): an open‐label, randomised phase 3 trial. Lancet Oncol. 2014;15:1269–1278. [DOI] [PubMed] [Google Scholar]
  • 170. Krupitskaya Y, Wakelee HA. Ramucirumab, a fully human mAb to the transmembrane signaling tyrosine kinase VEGFR‐2 for the potential treatment of cancer. Curr Opin Investig Drugs. 2009;10:597–605. [PubMed] [Google Scholar]
  • 171. Spratlin JL, Cohen RB, Eadens M, et al. Phase I pharmacologic and biologic study of ramucirumab (IMC‐1121B), a fully human immunoglobulin G(1) monoclonal antibody targeting the vascular endothelial growth factor receptor‐2. J Clin Oncol. 2010;28:780–787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172. LoRusso PM, Weiss D, Guardino E, Girish S, Sliwkowski MX. Trastuzumab emtansine: a unique antibody‐drug conjugate in development for human epidermal growth factor receptor 2‐positive cancer. Clin Cancer Res. 2011;17:6437–6447. [DOI] [PubMed] [Google Scholar]
  • 173. Corrigan PA, Cicci TA, Auten JJ, Lowe DK. Ado‐trastuzumab emtansine: a HER2‐positive targeted antibody‐drug conjugate. Ann Pharmacother. 2014;48:1484–1493. [DOI] [PubMed] [Google Scholar]
  • 174. Chari RVJ, Miller ML, Widdison WC. Antibody‐drug conjugates: an emerging concept in cancer therapy. Angew Chem Int Ed Engl. 2014;53:3796–3827. [DOI] [PubMed] [Google Scholar]
  • 175. Guerin M, Sabatier R, Goncalves A. Trastuzumab emtansine (Kadcyla (R)) approval in HER2‐positive metastatic breast cancers. Bull Cancer. 2015;102:390–397. [DOI] [PubMed] [Google Scholar]
  • 176. Michel LL, Bermejo JL, Gondos A, Marme F, Schneeweiss A. T‐DM1 as a new treatment option for patients with metastatic her2‐positive breast cancer in clinical practice. Anticancer Res. 2015;35:5085–5090. [PubMed] [Google Scholar]
  • 177. Lambert JM, Chari RVJ. Ado‐trastuzumab emtansine (T‐DM1): an antibody‐drug conjugate (ADC) for HER2‐positive breast cancer. J Med Chem. 2014;57:6949–6964. [DOI] [PubMed] [Google Scholar]
  • 178. Chudasama VL, Stark FS, Harrold JM, et al. Semi‐mechanistic population pharmacokinetic model of multivalent trastuzumab emtansine in patients with metastatic breast cancer. Clin Pharmacol Ther. 2012;92:520–527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179. Peddi PF, Hurvitz SA. Ado‐trastuzumab emtansine (T‐DM1) in human epidermal growth factor receptor 2 (HER2)‐positive metastatic breast cancer: latest evidence and clinical potential. Ther Adv Med Oncol. 2014;6:202–209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180. Patel KC, Hageman K, Cooper MR. Ado‐trastuzumab emtansine for the treatment of human epidermal growth factor receptor 2‐positive metastatic breast cancer. Am J Health Syst Pharm. 2014;71:537–548. [DOI] [PubMed] [Google Scholar]
  • 181. Burris HAI, Rugo HS, Vukelja SJ, et al. Phase II study of the antibody drug conjugate trastuzumab‐dm1 for the treatment of human epidermal growth factor receptor 2 (HER2) ‐positive breast cancer after prior HER2‐directed therapy. J Clin Oncol. 2011;29:398–405. [DOI] [PubMed] [Google Scholar]
  • 182. Hurvitz SA, Dirix L, Kocsis J, et al. Phase II randomized study of trastuzumab emtansine versus trastuzumab plus docetaxel in patients with human epidermal growth factor receptor 2‐positive metastatic breast cancer. J Clin Oncol. 2013;31:1157–1163. [DOI] [PubMed] [Google Scholar]
  • 183. Krop IE, LoRusso P, Miller KD, et al. A phase II study of trastuzumab emtansine in patients with human epidermal growth factor receptor 2‐positive metastatic breast cancer who were previously treated with trastuzumab, lapatinib, an anthracycline, a taxane, and capecitabine. J Clin Oncol. 2012;30:3234–3241. [DOI] [PubMed] [Google Scholar]
  • 184. Aapro MS. Denosumab for bone metastases from breast cancer: a new therapy option? J Clin Oncol. 2011;29:E419–E420. [DOI] [PubMed] [Google Scholar]
  • 185. Drooger JC, van der Padt A, Sleijfer S, Jager A. Denosumab in breast cancer treatment. Eur J Pharmacol. 2013;717:12–19. [DOI] [PubMed] [Google Scholar]
  • 186. Bekker PJ, Holloway DL, Rasmussen AS, et al. A single‐dose placebo‐controlled study of AMG 162, a fully human monoclonal antibody to RANKL, in postmenopausal women. J Bone Miner Res. 2004;19:1059–1066. [DOI] [PubMed] [Google Scholar]
  • 187. Fenech J, Macpherson I, Rizwanullah M, Lumsden G, Alfayez M. Denosumab‐induced hypocalcaemia in metastatic breast cancer. Breast. 2015;241:S81. [Google Scholar]
  • 188. Stopeck AT, Lipton A, Body J, et al. Denosumab compared with zoledronic acid for the treatment of bone metastases in patients with advanced breast cancer: a randomized, double‐blind study. J Clin Oncol. 2010;28:5132–5139. [DOI] [PubMed] [Google Scholar]
  • 189. Henry DH, Costa L, Goldwasser F, et al. Randomized, double‐blind study of denosumab versus zoledronic acid in the treatment of bone metastases in patients with advanced cancer (excluding breast and prostate cancer) or multiple myeloma. J Clin Oncol. 2011;29:1125–1132. [DOI] [PubMed] [Google Scholar]
  • 190. Cleeland CS, Body J, Stopeck A, et al. Pain outcomes in patients with advanced breast cancer and bone metastases results from a randomized, double‐blind study of denosumab and zoledronic acid. Cancer. 2013;119:832–838. [DOI] [PubMed] [Google Scholar]
  • 191. Verheul HMW, Hammers H, van Erp K, et al. Vascular endothelial growth factor trap blocks tumor growth, metastasis formation, and vascular leakage in an orthotopic murine renal cell cancer model. Clin Cancer Res. 2007;13:4201–4208. [DOI] [PubMed] [Google Scholar]
  • 192. Freyer G, Isambert N, You B, et al. Phase I dose‐escalation study of aflibercept in combination with docetaxel and cisplatin in patients with advanced solid tumours. Br J Cancer. 2012;107:598–603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193. Isambert N, Freyer G, Zanetta S, et al. Phase I dose‐escalation study of intravenous aflibercept in combination with docetaxel in patients with advanced solid tumors. Clin Cancer Res. 2012;18:1743–1750. [DOI] [PubMed] [Google Scholar]
  • 194. Chu QS. Aflibercept (AVE0005): an alternative strategy for inhibiting tumour angiogenesis by vascular endothelial growth factors. Expert Opin Biol Ther. 2009;9:263–271. [DOI] [PubMed] [Google Scholar]
  • 195. Sideras K, Dueck AC, Hobday TJ, et al. North central cancer treatment group (NCCTG) N0537: phase II trial of vegf‐trap in patients with metastatic breast cancer previously treated with an anthracycline and/or a taxane. Clin Breast Cancer. 2012;12:387–391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196. Tibbitts J, Cavagnaro JA, Haller CA, Marafino B, Andrews PA, Sullivan JT. Practical approaches to dose selection for first‐in‐human clinical trials with novel biopharmaceuticals. Regul Toxicol Pharmacol. 2010;58:243–251. [DOI] [PubMed] [Google Scholar]
  • 197. Selen A, Dickinson PA, Mullertz A, et al. The biopharmaceutics risk assessment roadmap for optimizing clinical drug product performance. J Pharm Sci. 2014;103:3377–3397. [DOI] [PubMed] [Google Scholar]
  • 198. Muller PY, Milton M, Lloyd P, Sims J, Brennan FR. The minimum anticipated biological effect level (MABEL) for selection of first human dose in clinical trials with monoclonal antibodies. Curr Opin Biotechnol. 2009;20:722–729. [DOI] [PubMed] [Google Scholar]
  • 199. Roque A, Lowe CR, Taipa MA. Antibodies and genetically engineered related molecules: production and purification. Biotechnol Prog. 2004;20:639–654. [DOI] [PubMed] [Google Scholar]
  • 200. Vicente T, Mota JPB, Peixoto C, Alves PM, Carrondo MJT, et al. Rational design and optimization of downstream processes of virus particles for biopharmaceutical applications: current advances. Biotechnol Adv. 2011;29:869–878. [DOI] [PubMed] [Google Scholar]
  • 201. Rathore AS, Agarwala H, Sharmaa AK, Pathaka M, Muthukumara S. Continuous processing for production of biopharmaceuticals. Prep Biochem Biotechnol. 2015;45:836–849. [DOI] [PubMed] [Google Scholar]
  • 202. Buyel JF, Fischer R. Downstream processing of biopharmaceutical proteins produced in plants the pros and cons of flocculants. Bioengineered. 2014;5:138–142. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Cell Proliferation are provided here courtesy of Wiley

RESOURCES