Molecular Targeted Therapy For Anticancer Treatment
Molecular Targeted Therapy For Anticancer Treatment
Molecular Targeted Therapy For Anticancer Treatment
com/emm
Since the initial clinical approval in the late 1990s and remarkable anticancer effects for certain types of cancer, molecular targeted
therapy utilizing small molecule agents or therapeutic monoclonal antibodies acting as signal transduction inhibitors has served as
a fundamental backbone in precision medicine for cancer treatment. These approaches are now used clinically as first-line therapy
for various types of human cancers. Compared to conventional chemotherapy, targeted therapeutic agents have efficient
anticancer effects with fewer side effects. However, the emergence of drug resistance is a major drawback of molecular targeted
therapy, and several strategies have been attempted to improve therapeutic efficacy by overcoming such resistance. Herein, we
summarize current knowledge regarding several targeted therapeutic agents, including classification, a brief biology of target
kinases, mechanisms of action, examples of clinically used targeted therapy, and perspectives for future development.
INTRODUCTION and aromatase inhibitors, which have been used for treatment of
Cancer is one of the main causes of disease-related death hormone receptor (HR)-dependent breast cancer and male and
worldwide. According to Global Cancer Observatory (GLOBOCAN) female reproductive cancers11; immune checkpoint inhibitors
estimates of cancer incidence and mortality, there were approxi- [e.g., antibodies against programmed cell death protein 1 (PD-1),
mately 19.3 million new cancer cases and almost 10.0 million programmed death-ligand 1 (PD-L1), or cytotoxic T-lymphocyte-
cancer deaths in 2020 globally1. The cancer-related burden (such associated protein 4 (CTLA-4)], which activate host antitumor
as incidence and mortality) is expected to be 28.4 million cases in immunity in a direct or indirect manner8,12; and even targeted
2040, which is a 47% increase compared with that in 2020, largely cytotoxic therapy that interferes with a specific cellular target
due to increases in risk factors, such as aging, socioeconomic (e.g., methotrexate, a dihydrofolate reductase inhibitor)10. Despite
development, overweight status, and smoking1,2. Therefore, it is the anticancer effectiveness of these targeted therapies, these
necessary to develop efficacious treatment strategies for patients drugs are only applicable for patients harboring targetable driver
with cancer. mutations or aberrations13,14. In addition, side effects or toxicity
Several therapeutic modalities, such as surgery, radiation caused by unexpected cross-reactivity with normal cells and
therapy, and systemic anticancer therapy, have been applied emergence of intrinsic or acquired drug resistance hamper their
clinically for cancer treatment, either alone, in combination, or effectiveness13,14. Notwithstanding some limitations, targeted
sequentially, depending on the stage, resectability, biology, therapy has resulted in remarkable survival benefits in some
comorbidities, and patient’s overall functional performance3,4. types of cancer and has led to a revolution in the fundamental
Systemic anticancer therapy, involving a wide range of anticancer concept of cancer treatment, providing the fundamental back-
drugs for treatment, palliation, symptom alleviation, and quality bone for evolution toward precision or personalized medicine in
of life improvement, includes cytotoxic chemotherapy, hormonal cancer13,15. Herein, we summarize current knowledge with
agents, targeted therapy, and antitumor immunotherapy5,6. respect to molecular targeted therapy, including the history,
Cytotoxic chemotherapy inhibits the survival of actively prolifer- types, and mechanism of action, and provide examples of
ating cells by disrupting the synthesis of DNA and RNA, blocking clinically available targeted therapy. In this paper, ‘targeted
mitosis, and/or forming covalent bonds with DNA, RNA, and therapy’ is confined to conventional molecular targeted therapy
proteins7, and it has been extensively used in adjuvant or (signal transduction inhibitors).
neoadjuvant therapy as well as in palliative therapy7. Due to the
disadvantages of chemotherapy, including side effects and Brief history of molecular targeted therapy
toxicity associated with nonselective action against actively Paul Ehrlich first proposed the concept of targeted therapy in the
proliferating normal cells2,8, there has been innovative develop- 1890s as a “magic bullet” that would be completely specific for the
ment of ‘targeted’ cancer treatment with increased cancer cell target and thus safe without any additional toxicity14,16. This
specificity8. Targeted therapy may include the following: conven- theory was initially applied to infectious diseases but not to
tional molecular targeted agents, such as small molecule anticancer therapy due to insufficient knowledge of the etiology
inhibitors or antibodies that specifically inhibit signal transduction and biology of cancer14,16; however, this concept has since been
pathways involved in growth, proliferation, and survival9,10; expanded to cancer treatment14,16. Trastuzumab, an anti-HER2
hormonal agents such as estrogen receptor (ER) antagonists monoclonal antibody, and imatinib, a small molecule tyrosine
College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea. ✉email: [email protected]
1
Fig. 1 Timeline for the approval of selected molecular targeted therapeutic agents. The first FDA-approved targeted therapeutic agent for
each cellular target (denoted in blankets) is indicated in the timeline.
kinase inhibitor targeting the BCR-ABL fusion-mediated aberrantly Mechanisms of the anticancer effects of molecular targeted
activated ABL kinase, were developed and clinically approved in therapy. Molecular targeted therapies achieve anticancer effects
1998 and 2001 for treatment of HER2-positive breast cancer and through various mechanisms, such as inhibition of cell prolifera-
Philadelphia chromosome-positive chronic myelogenous leuke- tion, metastasis, and angiogenesis, induction of apoptosis, and
mia, respectively14,17–19. The success of imatinib in the clinic has reversal of multidrug resistance2 (Fig. 2a). Several molecular
served as the paradigm for extensive use of small molecule kinase targeted therapeutic agents also facilitate host antitumor immu-
inhibitors as anticancer therapy8,17, and a number of anticancer nity by potentiating CD8+ T-cell recruitment and natural killer cell
molecular targeted therapies have been approved for clinical use cytotoxicity, downregulating immunosuppressive myeloid cells,
in cancer patients8,17. The timeline for the development of the and inducing immunogenic cell death, either alone or in
main molecular targeted therapy is illustrated in Fig. 1. combination with chemotherapeutic agents29. Therapeutic mAbs
create a bridge between tumor cells and immune cells via Fab
Types, mechanisms of action and resistance, and adverse region-mediated binding to a target protein of tumor cells and
effects/toxicity of molecular targeted therapy recognition of immune cells through the Fc region of antibodies30,
To date, numerous molecular targeted therapeutic agents have resulting in opsonization and antibody-dependent cellular cyto-
been used clinically for cancer treatment. The classification of toxicity (ADCC) toward tumor cells30 (Fig. 2b). A recent study
molecular targeted therapeutic agents and their targets, mechan- demonstrated that neutrophils mediate trogoptosis (Fig. 2c), the
ism of action, side effects, and toxicity are described below. phenomenon of transferring surface molecules of interacting cells
onto immune cells31,32, which causes lytic/necrotic death of
Types of molecular targeted therapy. The two major types of antibody-opsonized cancer cells33. mAbs and SMIs also exert
molecular targeted therapy are monoclonal antibodies (mAbs) immune cell-induced cytotoxic effects on cancer cells by
and small molecule kinase inhibitors (SMKIs)8,14. mAbs target activating complement and complement-dependent cytotoxi-
extracellular ligands (e.g., bevacizumab targets vascular endothe- city30,34, facilitating antigen processing by increasing expression
lial growth factor [VEGF]), membrane receptors (e.g., trastuzumab of major histocompatibility complex molecules30,35,36 and regulat-
targets HER2 and cetuximab; panitumumab targets EGFR), and ing cytokine/chemokine expression30,37.
membrane-bound proteins (e.g., rituximab targets CD20), acting
through ligand-binding blockade, ligand‒receptor interaction Mechanisms underlying resistance to molecular targeted therapy.
neutralization, or target molecule internalization/degradation14,20. The emergence of drug resistance is a major hurdle of efficacious
Except for inhibitors targeting nonkinase cellular proteins (e.g., anticancer treatment. Primary (intrinsic) resistance is defined as a
mutated KRAS and proteasome) or epigenetic modulators (e.g., refractory status to initial therapy due to intrinsic cellular, genetic,
histone deacetylases), most SMKIs suppress protein kinases and/or epigenetic alterations. Hyperactivation of compensatory
involved in the transformation, growth, proliferation, and survival signaling pathways [e.g., truncated HER2 expression (p95HER2) for
of cancer cells. As deregulation of protein kinases (e.g., activation resistance to anti-HER2 mAbs38; KRAS mutation or MET amplifica-
by gain-of-function genetic mutation, gene amplification, auton- tion for resistance to anti-EGFR therapy38,39], mutations in kinase
omous activation, and chromosomal rearrangement) has been domains (e.g., EGFR exon 20 insertion for resistance to anti-EGFR
associated with cancer development and progression21–24, protein therapy38), isoform switching (e.g., BRAF/CRAF switching for
kinases have been regarded as important targets for developing resistance to anti-BRAF therapy40), and metabolic reprogram-
molecular targeted therapies. Protein kinases are classified into ming40 during disease development are involved in primary
receptor tyrosine kinases, nonreceptor (cytoplasmic) tyrosine resistance to molecular targeted therapy.
kinases, serine/threonine kinases, and lipid kinases based on their Human cancers often exhibit substantial intratumor hetero-
subcellular localization, substrate type, and hallmark roles in geneity, which is a main driver for emerging acquired therapy
cancer21 (Fig. 2). A detailed explanation of the signal transduction resistance as a result of expansion of rare preexisting refractory
by receptor tyrosine kinase is described in previous studies24,25. populations during treatment in initial responders39,41,42. Various
SMKIs block the enzymatic activity of the aforementioned molecular and cellular alterations [e.g., development of second-
kinases via several modes of action26. Type I kinase inhibitors bind ary mutations [EGFR T790M and C797S38,43,44, BCR-ABL T315I44,
to the ATP-binding pocket of the active conformation of the BRAF V600E40,44, Bruton’s tyrosine kinase (BTK) C418S44, ana-
enzyme [DFG (Asp-Phe-Gly)-in and αC-helix-in]26, whereas type I1/2 plastic lymphoma kinase (ALK) G1202R, and ROS1 G2032R and
or type II inhibitors bind the enzyme in an inactive conformation D2033N44], alterations in noncoding RNAs44, activation of
(type I1/2: DFG-Asp in; type II: DFG-Asp out)21,26. Type III and type IV bypassing signaling pathways, including MET, HER2, type I
inhibitors allosterically suppress kinase activity by binding either insulin-like growth factor receptor (IGF-1R), and AXL43,45,
to a site next to the ATP-binding pocket or one remote from the mutations in BRAF, PTEN, PIK3CA, and MAP2K143,45, interaction
ATP-binding pocket located in the kinase substrate-binding with stromal cells in the tumor microenvironment43,46, altera-
site21,26,27. Type V inhibitors act as bivalent inhibitors binding to tions in E3 ubiquitin ligases47, reactivation of developmental
two different portions of the kinase lobe21,26. Type VI inhibitors processes, such as the epithelial-mesenchymal transition (EMT),
covalently bind an enzyme to inhibit kinase activity26,28. A recent acquisition of cancer stem cell (CSC)-associated phenotypes, and
paper describes the detailed mode of action of each type of kinase transdifferentiation to small-cell lung cancer43,48] have also been
inhibitor26, and some examples are listed in Table 1. shown to induce acquired therapy resistance. The mechanisms
Fig. 2 Mechanism of the anticancer effect of molecular targeted therapy. a Schematic diagrams of the main protumor signal transduction
pathways and their inhibition by molecular targeted therapeutic agents. b, c Schematic diagrams for antibody-dependent cellular cytotoxicity
b and trogoptosis c. See the text and relevant references for details.
of resistance to each molecular targeted therapy are summarized have indicated that cancer patients who receive targeted therapy
in Tables 2–6. may experience various side effects and toxicity. The side effects
of targeted therapy include asthenia, anorexia, dyspnea, diarrhea,
Adverse effects and toxicity of molecular targeted therapy. Despite nausea, vomiting, mucositis, skin rash, fever, hand-foot syn-
improved specificity for cancer cells, epidemiological studies drome, fatigue, cardiotoxicity, hypertension, and bleeding49,50.
EGFR Panitumumab Vectibix Metastatic CRC (2006) RAS/BRAF mutation Integument toxicity,
(Abgenix/Amgen) MET amplification skin toxicity, diarrhea
PTEN loss
79,80
EGFR Amivantamab Rybrevant Advanced NSCLC with EGFR Infusion reactions
(JNJ-61186372) (Janssen Biotech) exon 20 insertion mutations ocular toxicity,
progressing after platinum- peripheral edema,
based chemotherapy (2021) hypoalbuminemia
79,81
EGFR Mobocertinib Exkivity Advanced NSCLC with EGFR Diarrhea, skin toxicity
(TAK-788) (Takeda exon 20 insertion mutations
Pharmaceuticals) progressing after platinum-
based chemotherapy (2021)
8,56,90,209
HER2 Trastuzumab Herceptin Metastatic breast Locally advanced Truncation of HER2 Cardiotoxicity
(Genentech/ cancer (1998) unresectable or metastatic extracellular domain
Roche) HER2+ gastric or (p95 HER2)
gastroesophageal junction PTEN loss
(GEJ) adenocarcinoma in IGF-1R expression
combination with PIK3CA mutation
pembrolizumab (2021)
1673
1674
Table 2. continued
Target Generic name Brand name First approved Additional indication Drug resistance Side effects/toxicity References
(Code name) (Company) indication (Year) mechanism (selected) (selected)
8,90,210
HER2 Pertuzumab Perjeta HER2+ early breast cancer Diarrhea, nausea,
(Genentech/ (EBC) with high risk of alopecia, fatigue,
Roche) recurrence (2017) peripheral neuropathy,
vomiting
91
HER2 Zanidatamab (Zymeworks) Advanced/metastatic HER2- Diarrhea, infusion-
(ZW25) expressing biliary tract cancers related reactions
82,83,209,211
HER2 Lapatinib Tykerb HER2+ metastatic breast Triple-positive metastatic Crosstalk with ER Diarrhea, skin rash,
(GW-572016) (GlaxoSmithKline/ cancer progressing with prior breast cancer (in HER2 mutation asymptomatic
Novartis) therapy (in combination with combination with PIK3CA mutation cardiotoxicity
capecitabine, 2007) letrozole, 2010) AXL elevation
HER2 L755S mutation
56,84,209,211,212
HER2 Neratinib Nerlynx Extended adjuvant therapy for Advanced or metastatic TORC1 hyperactivation Diarrhea
(HKI-272) (Puma HER2+ breast cancer (2017) HER2+ breast cancer RAS upregulation
Biotechnology) progressing with prior
therapy (in combination
with capecitabine, 2020)
85,211
HER2 Tucatinib Tukysa Advanced or metastatic HER2+ HER2 L755S mutation Diarrhea
(ONT-380) (Seattle Genetics) breast cancer (in combination cardiotoxicity
with trastuzumab and
capecitabine, 2020)
79,96,98,99,213
ALK Crizotinib Xalkori Locally advanced or metastatic ROS1-positive ALK mutation (G1269A, Nausea, vomiting,
ROS1 (PF-02341066) (Pfizer) ALK+ NSCLC (2011) NSCLC (2016) C1156Y, E1210K, diarrhea, visual
MET I1171T, S1206C/Y, disturbance, sinus
H.-Y. Min and H.-Y. Lee
imatinib (2020)
91,224,225
FGFR Erdafitinib Balversa Metastatic urothelial Metastatic or locally FGFR1 V561M/F Hyperphosphatemia,
(JNJ‑42756493) (Janssen cancer (2018) advanced bladder cancer mutation dry mouth, diarrhea,
Pharmaceuticals) with an FGFR3 or FGFR2 FGFR2 N549H fatigue, stomatitis
alteration (2019) mutation
p.E565A and p.L617M
single-nucleotide
variants
91,224–226
FGFRs Pemigatinib Pemazyre Previously treated, FGFR1 V561M/F Hyperphosphatemia,
(INCB054828) (Incyte unresectable, locally mutation dry mouth, diarrhea,
Corporation) advanced, or metastatic FGFR2 N549H fatigue, stomatitis
cholangiocarcinoma with mutation
FGFR2 fusion or other
rearrangements (2020)
91,224,225
FGFRs Futibatinib (Taiho Locally advanced/metastatic p.E565A and p.L617M Hyperphosphatemia,
(TAS-120) Pharmaceutical) cholangiocarcinoma with single-nucleotide dry mouth, diarrhea,
FGFR2 gene variants paronychia,
rearrangement (2021) FGFR2 V564F mutation
91,224,225
FGFRs Infigratinib Truseltriq Locally advanced/metastatic FGFR2 N549H, N550H/ Hyperphosphatemia,
(BGJ398) (QED cholangiocarcinoma with K, V564F, E565A, dry mouth, diarrhea,
Therapeutics FGFR2 gene K660M, L618V, K641R fatigue, stomatitis
/Helsinn) rearrangement (2021) mutation
91,224,225
FGFRs Derazantinib (Basilea Intrahepatic Hyperphosphatemia,
(ARQ 087) Pharmaceutica cholangiocarcinoma (2021) dry mouth, diarrhea,
/Merck) fatigue, stomatitis
227,228
effects are related to therapy response52. Severe toxicities, such
as colitis, digestive perforation, toxic cardiomyopathy, pneumo-
nitis/interstitial lung disease, acute respiratory distress syndrome,
anemia, hypertension posterior reversible encephalopathy syndrome, necrotizing
fasciitis, acute renal failure, and hypersensitivity, have been
Side effects/toxicity
AST/ALT elevation,
AST/ALT elevation,
RET mutation at
RET mutation at
G810, L730
NSCLC (2020)
Medicines)
Oncology)
(Blueprint
Retevmo
RET
A337, W464,
P465, V468, I502
151,152,235,236
BTK Ibrutinib Imbruvica MCL7 (2013) CML (2014) BTK C481S, Atrial fibrillation, bleeding,
(PCI-32765) (Pharmacyclics/ Waldenström’s T474I/M hypertension, diarrhea,
AbbVie/Janssen) Macroglobulinemia (2015) mutation nausea, vomiting
CLL8 (first line) and SLL9 (2016)
Relapsed/refractory MZL10 (2017)
151,152,235,236
BTK Acalabrutinib Calquence Relapsed/refractory Relapsed/refractory CLL (2019) BTK C481S Atrial fibrillation, bleeding,
(ACP-196) (Acerta Pharma/ MCL (2017) mutation hypertension, diarrhea,
AstraZeneca) nausea, vomiting
151,152,235,236
BTK Zanubrutinib Brukinsa MCL (2019) Waldenström’s BTK C481S Diarrhea, nausea,
(BGB-3111) (BeiGene) Macroglobulinemia (2021) mutation vomiting
Relapsed/refractory MZL (2021)
154,155,237
JAK Ruxolitinib Jakafi Myelofibrosis (2011) Cytopenia, diarrhea,
(INC424) (Incyte/Novartis) nausea, vomiting
1681
H.-Y. Min and H.-Y. Lee
1682
ephrin receptor EPHB4138. Bosutinib is an orally active and ATP-
References competitive dual SFK/ABL inhibitor135,137,138 showing similar
inhibitory effects against mutated or amplified BCR-ABL associated
154,155,237
with imatinib resistance137,140 and BCR-ABL harboring the T315I
mutation137,140. Accordingly, bosutinib has been used for treat-
ment of patients with Ph+ CML who are resistant to or intolerant
of imatinib141. Other agents approved in the clinic include
radotinib, an orally active second-generation BCR-ABL inhibitor
that exhibits inhibitory effects on wild-type and some imatinib-
resistant mutant forms of BCR-ABL and PDGFR8,142, and asciminib,
Side effects/toxicity
Src-family kinases (SFK: Blk, Fgr, Frk, Fyn, Hck, Lck, Lyn, Src, Yes,
and Yrk) contain a conserved domain organization consisting of a
myristoylated N-terminal segment (SH4 domain), followed by SH3,
SH2, linker, and tyrosine kinase domains and a short C-terminal
tail146,147. Among SFKs, Src, Fyn, and Yes are ubiquitously
expressed; Hck, Lck, Lyn, Blk, Yrk, and Fgr are primarily expressed
in hematopoietic cells and Frk-related kinases in epithelial-derived
tissues. Similar to ABL, SFKs adopt an inactive conformation
through autoinhibitory intramolecular interactions involving
Additional indication
and stability of the protein, SH3 and SH2 domains, and the catalytic
ALL: acute lymphocytic leukemia.
JAK
BRAF V600E-mutant
anaplastic thyroid cancer
in combination with
trametinib (2018)
157,167,168,239–241
BRAF Encorafenib Braftovi Unresectable or metastatic melanoma BRAF V600E-mutant NRAS mutation Rash, diarrhea,
(LGX818) (Novartis/Array with BRAF mutations in combination metastatic CRC2 in CRAF fatigue, arthralgia
BioPharma) with binimetinib (2018) combination with overexpression
cetuximab (2020) secondary BRAF
mutation
MEK1/2
mutation
157,167,168,239–241
MEK Trametinib Mekinist BRAF V600E-mutant advanced Advanced melanoma RTK reactivation Rash, diarrhea,
(GSK1120212, (GlaxoSmithKline/ melanoma (2013) with BRAF mutation in PI3K, STAT3 fatigue, arthralgia
JTP-74057) Novartis) combination with activation
dabrafenib (2014)
BRAF V600E-mutant
metastatic NSCLC1 in
combination with
dabrafenib (2017)
BRAF V600E-mutant
anaplastic thyroid cancer
in combination with
dabrafenib (2018)
157,167,168,239–242
MEK Cobimetinib Cotellic Advanced melanoma with BRAF RTK reactivation Rash, diarrhea,
(GDC-0973, (Genentech) mutation in combination with PI3K/STAT3 fatigue, arthralgia
RG7420) vemurafenib (2015) activation
mTOR Temsirolimus Torisel Advanced RCC6 (2007) RTK reactivation Hyperglycemia, rash,
(CCI-779) (Pfizer) stomatitis, fatigue,
nausea, diarrhea
171,172,243,244
mTOR Everolimus Afinitor RCC after failure of sunitinib or sorafenib Advanced pancreatic RTK reactivation Hyperglycemia, rash,
(RAD001) (Novartis) treatment (2009) neuroendocrine stomatitis, fatigue,
tumor (2011) nausea, diarrhea
HR+ and HER2- breast
cancer for use in
combination with
exemestane (2012)
Subependymal giant-cell
astrocytoma (2012)
8,173,245
CDK4/6 Palbociclib Ibrance Advanced or metastatic breast HR+ and HER2- CDK4/6 Fatigue, nausea,
(PD 0332991) (Pfizer) cancer (2015) metastatic breast overexpression diarrhea, vomiting
cancer (2016)
8,174,245
CDK4/6 Ribociclib Kisqali HR+ and HER2- metastatic breast CDK4/6 Fatigue, nausea,
(LEE011) (Novartis) cancer (2017) overexpression diarrhea, vomiting
1685
H.-Y. Min and H.-Y. Lee
1686
dissociates from DNA, and the DNA repair process is completed by
References recruitment of DNA repair proteins176. BReast CAncer gene 1
(BRCA1) and BRCA2 (BRCA1/2) are tumor-suppressor genes that
8,245,246
play a key role in repair of double-strand DNA breaks via the
conservative homologous recombination repair (HRR) pro-
cess175,177. Mutations in BRCA1/2 genes have been found in
some cancer types, including breast, ovarian, pancreatic, and
prostate cancers177. Defects in BRCA function due to BRCA1/2
Side effects/toxicity
diarrhea, vomiting
gene mutations cause loss of the HRR process and mediate the
Fatigue, nausea,
overexpression
drug resistance.
Because the side effects and toxicity of targeted therapy are
Generic name
(Code name)