Download as pdf or txt
Download as pdf or txt
You are on page 1of 7

Chinese Journal of Chemical Engineering 30 (2021) 301–307

Contents lists available at ScienceDirect

Chinese Journal of Chemical Engineering


journal homepage: www.elsevier.com/locate/CJChE

Review

Monoclonal antibody-based cancer therapies


Yingnan Si 1, Arin L. Melkonian 2, Keegan C. Curry 1, Yuanxin Xu 1, Maranda Tidwell 1,
Mingming Liu 3, Ahmed F. Zaky 4, Xiaoguang (Margaret) Liu 1,⇑
1
Department of Biomedical Engineering, University of Alabama at Birmingham (UAB), 1825 University Blvd, Birmingham, AL 35294, USA
2
Department of Medicine, UAB, 1720 2nd Ave S., Birmingham, AL 35294, USA
3
Chinese Medicine Hospital, 98 Shengcheng Street, Shouguang 262700, China
4
Department of Anesthesiology and Perioperative Medicine, UAB, 625 19th Street South, Birmingham, AL 35294, USA

a r t i c l e i n f o a b s t r a c t

Article history: Targeted therapy has been widely demonstrated as an effective strategy to treat cancers, the leading
Received 13 October 2020 cause of death in the world. This minireview summarizes the technical platforms and methodologies uti-
Received in revised form 15 November 2020 lized to develop and engineer therapeutic monoclonal antibodies and antibody-drug conjugates. First, the
Accepted 16 November 2020
USA FDA approved monoclonal antibody (mAb)-based targeted therapies are reviewed. Then the repre-
Available online 27 November 2020
sentative innovative chimeric, humanized and fully human anti-cancer antibodies and antibody-drug
conjugates are described. Finally, the past and predictive market trend of therapeutic antibodies is
Keywords:
discussed.
Monoclonal antibody
Antibody-drug conjugate
Ó 2020 The Chemical Industry and Engineering Society of China, and Chemical Industry Press Co., Ltd. All
Anti-cancer therapy rights reserved.
Market

1. Introduction To develop therapeutic mAbs, hybridoma technology, which


fuses the antibody producing mouse immunized B spleen cells
Despite the advances in research, diagnosis, and treatment, can- and reproducing myeloma (an immortal cancer B cells), was estab-
cer remains one of the leading causes of death worldwide, with an lished by Köhler and Milstein in 1975 [15–17]. The advantages of
estimated 9.6 million deaths in 2018 [1] and an expected rise to hybridoma-based mAbs are the high antigen binding and low
29.5 million new cases and 16.4 million deaths by 2040 (Interna- aggregation in vivo [18–22]; however, the generated murine mAbs
tional Agency for Research on cancer). The major obstacles have short half-lives, low biological activity, and reduced initiation
involved in preventing the success and efficacy of standard of effector function [23,24]. In 1985, Smith et al. developed phage
chemotherapies include limited clinical efficacy, development of display technology to develop mAb by screening a pre-
drug resistance, severe off-target side effects, and recurrence post established library of mouse or human antibodies [25,26]. Phage
primary treatment. With the intensive efforts for discovering the display screening is fast but the developed mAbs have low antigen
cancer-specific antigens or signaling pathways such as HER2, EGFR, targeting specificity, sensitivity, and affinity.
CD20, CD22, CD30, CD33, CD38, CD52, CTLA-4, PD-L1, and others Antibody engineering is a powerful tool to develop novel ther-
[2–6], numerous targeted anti-cancer strategies that overcome apeutic mAbs for clinical application. For example, after sequenc-
these barriers, such as monoclonal antibody (mAb)-based biologics ing and cloning, murine mAbs can be further engineered to
[7,8], have been developed or are being evaluated in clinical trials. generate either chimeric, humanized, full human, or bispecific
The mAbs have been demonstrated as effective biopharmaceuticals antibodies [7,27–31]. The first humanized mAb (anti-IL2 daclizu-
that can improve the overall survival of cancer patients via multi- mab) was developed by grafting the complementary-determining
ple anti-cancer mechanisms, including inhibition of cell prolifera- region (CDR) of murine antibody with the human mAb framework
tion, induction of apoptosis, antibody-dependent cell-mediated sequence and was approved in 1997 by the US FDA [32]. The trans-
cytotoxicity (ADCC) [9–12] and complement dependent cytotoxic- genic animal technology [33] and phage display [34] enabled the
ity (CDC) [13,14] (Fig. 1). development of fully human mAb. The first human antibody, pan-
itumumab generated from XenoMouse transgenic mice, treats epi-
⇑ Corresponding author at: Department of Biomedical Engineering, University of
dermal growth factor receptor (EGFR) positive metastatic
Alabama at Birmingham, 1825 University Blvd., Birmingham, AL 35294, USA. Fax:
colorectal cancer and was approved by the US FDA in 2006 [35].
+1 205 996 4701. Compared to murine mAb, the constant region in humanized and
E-mail address: mliu@uab.edu (Xiaoguang (Margaret) Liu). human mAbs reduces immunogenicity, improves effector

https://doi.org/10.1016/j.cjche.2020.11.009
1004-9541/Ó 2020 The Chemical Industry and Engineering Society of China, and Chemical Industry Press Co., Ltd. All rights reserved.
302 Y. Si et al. / Chinese Journal of Chemical Engineering 30 (2021) 301–307

Fig. 1. The anti-cancer mechanisms of mAb and ADC. 1. mAb or ADC specifically targets cancer cells by binding the overexpressed surface receptor; 2. ADC internalizes
mediated with receptor; 3.1. Released cytotoxic drug causes cancer cell death; and 3.2. mAb-mediated ADCC or CDC leads to cytolysis.

functions, and significantly extends the serum half-life [36]. More- 3. Engineering of Therapeutic mAb
over, the affinity maturation, effector function modification, and
pharmacokinetic modulation technologies have been developed Immunogenicity is one of the major factors that hinders the
to improve the stability and function of mAbs [27,37]. therapeutic application of murine mAb. For instance, obvious
Since the FDA approved the first anti-cancer chimeric mAb in human anti-murine antibody responses in the circulation can be
1997, anti-CD20 Rituximab, a treatment for non-Hodgkin lym- detected within one to three weeks post-murine mAb administra-
phoma [38,39], 32 more mAbs have been approved for cancer tion and the serum half-life of murine mAb is only 15–30 h [56,57].
treatment in the USA [40,41] (Table 1). The CD19-targeting Although chimeric and humanized mAbs have been successfully
Tafasitamab-cxix and anti-TROP-2 Sacituzumab govitecan were developed, their immunogenicity is still challenging [58–60].
approved in 2020 for the treatment of adult patients with relapsed Phage display [61–63] and transgenic mice [64–68] have enabled
or refractory diffuse large B-cell lymphoma and triple-negative the development of clinically indistinguishable mAbs (Table 1).
breast cancer, respectively. There are five additional new antibody To date, over 500 therapeutic mAbs have been evaluated in clinical
therapeutics undergoing FDA review in 2020 [8]. In addition to trials worldwide, with more than 80 mAbs (32 of them for cancer
cancer treatment, the mAbs can also be used to diagnose cancers, treatment) approved by the US FDA for clinical use (Fig. 3). Here we
construct antibody-drug conjugate, or direct the targeted delivery use representative FDA-approved mAbs to review a multitude of
of various therapies [42–47]. advanced antibody engineering technologies.

2. Antibody Structure
3.1. Cetuximab (chimeric antibody)
Of the five classes of immunoglobin family (IgA, IgD, IgE, IgM,
and IgG), IgG is the most effective antibody for cancer treatment A chimeric antibody can be generated by fusing the variable
[48,49]. IgG is comprised of two identical heavy chains (HC, region of a murine antibody with the constant region of a human
50000) and two identical light chains (LC, 25000) that are held antibody, containing the consensus sequences for N-linked glyco-
together by four interchain disulfide bonds and arranged in a Y sylation and the N-terminal amino acid cyclized as pyroglutamic
shape (Fig. 2A) [50,51]. The HC consists of one variable domain acid [7,27–31,69]. Cetuximab (Erbitux) is a 152000 chimeric mAb
(VH), a discrete folded region containing 110 amino acids, three containing a human IgG1 constant region and a murine Fv variable
constant domains (CH1, CH2 and CH3), and a hinge linker. The region, which was developed to treat metastatic colorectal cancer,
LC consists of one variable domain (VL) and constant domain metastatic non-small cell lung cancer, and head and neck cancer by
(CL) with two subtypes, lambda (k) and kappa (j). The domains targeting epidermal growth factor receptor (EGFR). Compared to
of VH, VL, CL and CH1 form the fragment antigen binding (Fab) murine antibodies, Cetuximab showed significantly improved
region which recognizes and binds a specific antigen. The CH2 anti-cancer efficacy, reduced immunogenicity, higher affinity to
and CH3 form the fragment crystallizable (Fc) region that mediates antigen, and reduced dosage [70–72]. For example, the combina-
the recruitment of immune cells through interaction with Fc tion of Cetuximab with platinum-based therapy using fluorouracil
gamma receptors (FccR) [52,53]. The four subclasses of IgG has resulted in overall survival (OS), progression-free survival
(IgG1-4) are highly conserved and differ in their hinge region (PFS), and objective response rate (ORR) of 10.1 months,
length, the number and location of interchain disulfide bonds, 5.5 months, and 35.6%, respectively, in the treatment of K-Ras
and the upper CH2 domains [51]. The Fc region of IgG subclasses wild-type and EGFR-positive head and neck cancer patients. In
leads to their functional difference in ADCC and CDC [40,54,55]. combination with the chemotherapeutic drug FOLFIRI, the Cetux-
Y. Si et al. / Chinese Journal of Chemical Engineering 30 (2021) 301–307 303

Table 1
Summary of commercialized mAb-based anti-cancer therapies.

Scientific/ Category Target Cancers Company Ref.


Brand name
Trastuzumab/ Humanized mAb HER2/ HER2-positive breast cancer, stomach cancer Genentech [42]
Herceptin neu (Roche)
Cetuximab/ Chimeric mAb EGFR Metastatic colorectal cancer, metastatic non-small cell lung cancer and head Eli Lilly and Co. [43,44]
Erbitux and neck cancer
Bevacizumab/ Humanized mAb VEGF Colorectal, lung, cervical, renal cell cancers, glioblastoma Genentech [4–6]
Avastin
Rituximab/ Chimeric mAb CD20 Non-Hodgkin’s lymphoma, chronic lymphocytic Leukemia IDEC/ [7,8]
Rituxan Genentech
Pertuzumab/ Humanized mAb HER2 HER2-positive breast cancer Genentech [9,10]
Perjeta
Alemtuzumab/ Humanized mAb CD52 Chronic lymphocytic leukemia Genzyme [11]
Campath
Ipilimumab/ Human mAb CTLA-4 Melanoma Bristol-Myers [12]
Yervoy Squibb
Atezolizumab/ Humanized mAb PD-L1 Urothelial carcinoma, non-small cell lung cancer Genentech [13,14]
Tecentriq
Gemtuzumab Humanized mAb-drug CD33 Acute myeloid leukemia Wyeth/Pfizer [15,16]
Ozogamicin/ conjugate
Mylotarg
Brentuximab Chimeric mAb-drug CD30 Hodgkin Lymphoma Seattle [17,18]
Vedotin/ conjugate Genetics
Adcentris
Nivolumab/ Human mAb PD-1 Melanoma, non-small cell lung cancer, Hodgkin lymphoma, renal cell Bristol-Myers [19]
Opdivo carcinoma Squibb
Elotuzumab/ Humanized mAb SLAMF7 Multiple myeloma Bristol-Myers [20]
Empliciti Squibb
Obinutuzumab/ Humanized mAb CD20 Chronic lymphocytic leukemia Genentech [21]
Gazyva
Daratumumab/ Humanized mAb CD38 Multiple myeloma Janssen Biotech [22,23]
Darzalex
Ibritumomab Mouse mAb CD20 Non-Hodgkin’s Lymphoma Spectrum [24]
Tiuxetan/ Pharm.
Zevalin
Ofatumumab/ Human mAb CD20 Chronic lymphocytic leukemia Glaxo [25]
Arzera
Necitumumab/ Human mAb EGFR squamous non-small cell lung cancer Eli Eilly [26]
Portrazza
Pembrolizumab/ Humanized mAb PD-1 melanoma, non-small cell lung, head and neck squamous cell, gastric, Merck [27]
Ketruda cervical, hepatocellular cancers,
Hodgkin lymphoma, urothelial carcinoma
Dinutuximab/ Chimeric mAb GD2 Neuroblastoma United [28]
Unituxin Therapeutics
Ado-Trastuzumab Humanized mAb HER2 HER2-positive breast cancer Genentech [29]
Emtansine/
Kadcyla
Tositumomab/ Murine mAb CD20 Non-Hodgkin’s Lymphoma Glaxo [30]
Bexxar
Ramucirumab/ Human mAb VEGFR2 Gastric Cancer Eli Lilly [31]
Cyramza
Avelumab/ Human mAb PD-L1 Merkel cell carcinoma EMD Serono [33]
Bavencio
Panitumumab/ Human mAb EGFR Colorectal Cancer Amgen [32]
Vectibix
Blinatumomab/ Murine mAb CD19, Acute Lymphoblastic Leukemia Amgen [34]
Blincyto CD3
Durvalumab/ Human mAb PD-L1 Urothelial Carcinoma AstraZeneca [35]
Imfinzi
Inotuzumab Humanized, drug CD22 Acute Lymphoblastic Leukemia Wyeth/Pfizer [36]
ozogamicin/ conjugate mAb
Besponsa

imab extended the OS, PFS, and ORR to 23.5 months, 9.5 months, meric antibody [7,27–31]. Trastuzumab (Herceptin) is a human-
and 57% from 19.6 months, 8.9 months, and 46% respectively [73]. ized mAb utilized to treat the HER2-positive metastatic breast
cancer, stomach cancer and gastroesophageal junction adenocarci-
3.2. Trastuzumab (humanized antibody) nomas. The anti-HER2 murine mAb was first developed through
hybridoma technology by immunizing mice with NIH 3T3 cells
A humanized antibody can be generated by substituting the that highly express human HER2 receptor. The hybridoma clone
CDR of a murine antibody with the corresponding CDR graft of a with high anti-HER2 mAb productivity and high HER2 selectivity
human antibody, which has a higher proportion of human regions was then screened. Finally, the variable region of mouse
(90%–94%) and more similarity to fully human antibody than chi- anti-HER2 antibody was grafted onto human antibody backbone,
304 Y. Si et al. / Chinese Journal of Chemical Engineering 30 (2021) 301–307

Fig. 2. The structures of mAbs. A. Overall structure of mAb; B. Structures of mouse, chimeric, humanized, and fully human mAbs.

cross linker to couple different mAbs or fragments at their hinge


cysteine residues [84–87], or recombinant DNA technologies (ge-
netic) such as knobs-into-holes, leucine zippers, minibodies, dia-
bodies, and single chain structures [30,31,82,83]. Compared to
traditional mAbs, BsAbs are advantageous in redirecting immune
cells to tumor cells to maximize tumor killing, simultaneously
blocking two different antigens to prevent activation of deleterious
downstream pathways, increasing binding affinity, and avoiding
potential antigen-loss relapse. Catumaxomab (Removab) is a bis-
pecific antibody containing two binding specificities towards the
epithelial cell adhesion molecule (EpCAM) and the T-cell antigen
CD3 [30,31,88,89]. This BsAb is produced in a mouse-rat quadroma
that contains one mouse IgG2a heavy-light chain and one rat IgG2b
heavy-light chain.

4. Antibody-drug Conjugate (ADC)

Fig. 3. The USA FDA approved therapeutic mAbs.


In addition to monotherapy, mAbs can be conjugated with
generating the humanized Trastuzumab [24,74–77]. Trastuzumab highly potent chemotherapies to construct antibody-drug conju-
can be used to treat breast cancer as a monotherapy or in combina- gates (ADCs) via the conjugation sites (e.g., free amines and thiols)
tion with the thermotherapy paclitaxel. and a chemical linker [90,91]. Linkers with high stability in acidic
and protease-rich environments both extracellularly and intracel-
3.3. Nivolumab (fully human antibody) lularly have been developed, such as acid labile, reducible,
enzyme-cleavable, and peptide-based linkers. As effective anti-
A fully human mAb can be generated by display technologies cancer biopharmaceuticals, ADCs integrate the advantages of
mAbs, which can specifically bind a tumor associated surface
[25,26], transgenic animals [35,36], and human B cells [78–80].
receptor and mediate cytotoxicity, with the potent cytotoxicity of
Nivolumab (Opdivo) is a PD-1-targeted IgG4 human antibody cre-
small molecule chemotherapeutics. The mAb enables an ADC cir-
ated from transgenic mice. In clinical applications, Nivolumab
culating through the bloodstream to bind the tumor specific sur-
treats patients with metastatic melanoma, metastatic squamous
face antigen. After binding, the ADC is internalized via receptor-
cell or squamous non-small-cell lung carcinoma, and advanced
mediated endocytosis, a late endosome is formed, and lysosomal
renal cell carcinoma as a monotherapy or in combination with Ipil-
degradation releases cytotoxic drug into the cytoplasm, ultimately
imumab (Yervoy) [80]. The combination of Nivolumab and Ipili-
leading to cancer cell death [92]. Multiple ADCs have been devel-
mumab improved the OS, PFS, and ORR in patients with
oped to effectively treat cancers in clinics while minimizing side
metastatic melanoma compared to Nivolumab alone.
effects in normal cells [93–99]. Here we use three FDA approved
ADCs as examples to describe the construction, anti-cancer mech-
3.4. Catumaxomab (bispecific antibody)
anisms, and clinical applications of this mAb-directed therapy.
A Bispecific Antibody (BsAb) can recognize two distinct antigen
targets or epitopes simultaneously. BsAbs can be constructed by 4.1. Gemtuzumab ozogamicin
employing quadroma technology (biological) that somatically
fuses two antibodies producing parent hybridomas into a hybrid- Gemtuzumab ozogamicin (Mylotarg) has been developed to
hybridoma cell [81–84], chemical conjugation that uses a chemical treat CD33-positive acute myeloid leukemia (AML) [100–106]. This
Y. Si et al. / Chinese Journal of Chemical Engineering 30 (2021) 301–307 305

ADC is composed of a humanized anti-CD33 IgG4 mAb (hP67.6) and achieve controlled drug release in tumor could further advance
and N-acetyl gamma calicheamicin, a DNA-binding cytotoxic the ADCs technologies.
antibiotic, combined through an acid-cleavable hydrazone linker.
The utilization of the lysine attachment site on the mAb can syn-
6. Conclusions
thesize active hydrazone conjugates without oxidation of the car-
bohydrates. After targeting the surface CD33 receptor and
mAb has evolved as an important biopharmaceutical in targeted
internalizing, the hydrazone linker is hydrolytically cleaved and
anti-cancer delivery as monotherapy or as a conjugated therapy.
the potent payload is released intracellularly. The released N-
Multiple platforms or technologies of therapeutic antibody devel-
acetyl gamma calicheamicin dimethyl hydrazide induces double-
opment, including hybridoma technology, phage display, and
stranded DNA breaks and further leads to the apoptosis of cancer
transgenic mice, have been developed. Various engineered anti-
cells. The clinical data demonstrated that the gemtuzumab
bodies, such as chimeric, humanized, and fully human antibodies,
ozogamicin treatment group achieved event-free survival (EFS),
have been developed to overcome the limitation of murine anti-
relapse-free survival (RFS) and OS of 40.8% (32.8–50.8), 50.3%
bodies and are widely used in clinics. The other antibody-
(41.0–61.6) and 53.2% (44.6–63.5), respectively, without severe
facilitated targeted therapies, such as antibody-drug conjugates
toxicity compared to EFS of 17.1% (10.8–27.1), RFS of 22.7%
and antibody tagged polymeric nanoparticles for drug delivery,
(14.5–35.7) and OS of 41.9% (33.1–53.1) in control group [107].
have also been rapidly developed.

4.2. Brentuximab vedotin


Declaration of Competing Interest
Brentuximab vedotin (Adcetris) is constructed by conjugating a
The authors declare that they have no known competing finan-
153-kDa chimeric anit-CD30 IgG1 antibody with monomethyl
cial interests or personal relationships that could have appeared
auristatin E via a protease-cleavable linker with a drug-antibody
to influence the work reported in this paper.
ratio of approximate 4 [108–111]. Brentuximab vedotin was devel-
oped to treat classical Hodgkin lymphoma, systemic anaplastic
large cell lymphoma, and any CD30-expressing peripheral T-cell Acknowledgements
lymphomas. The mAb-delivered monomethyl auristatin E induces
cancer cell apoptosis and cell death by binding the microtubule This work was supported by National Institutes of Health
and disrupting its networks. Significant improvements in objective R21CA226491-01A1 (X.M.L.), 1R01CA238273-01A1 (X.M.L.) and
response and survival benefit were observed in brentuximab vedo- 1R01CA242917-01A1 (X.M.L.).
tin treated patients compared to conventional therapies [112]. For
instance, a Phase III clinical study with 128 enrolled patients References
showed that the objective response rate lasting at least 4 months
(ORR4) was 56.3% in treatment group versus 12.5% in the conven- [1] F. Bray, J. Ferlay, I. Soerjomataram, R.L. Siegel, L.A. Torre, A. Jemal, Global
tional physician’s choice group. The brentuximab vedotin also cancer statistics 2018: GLOBOCAN estimates of incidence and mortality
worldwide for 36 cancers in 185 countries, CA Cancer J. Clin. 68 (2018) 394–
extended the median progression-free survival to 17.2 months 424.
from 3.5 months in control group. [2] E.V. Stevens, E.M. Posadas, B. Davidson, E.C. Kohn, Proteomics in cancer, Ann.
Oncol. Off. J. Eur. Soc. Med. Oncol./ESMO 15 Suppl 4 (2004), iv167–171.
[3] D. Rosenblum, D. Peer, Omics-based nanomedicine: the future of personalized
4.3. Trastuzumab emtansine oncology, Cancer Lett. 352 (2014) 126–136.
[4] D.F. Hayes, OMICS-based personalized oncology: if it is worth doing, it is
worth doing well!, BMC Med. 11 (2013) 221–224.
Trastuzumab emtansine (Kadcyla) is constructed by conjugat- [5] W.J. Rettig, L.J. Old, Immunogenetics of human cell surface differentiation,
ing a humanized anti-HER2 IgG1 mAb with a microtubule inhibi- Annu. Rev. Immunol. 7 (1989) 481–511.
tory mertansine by a thioether chemical linker with a drug- [6] D.T. Loo, J.P. Mather, Antibody-based identification of cell surface antigens:
targets for cancer therapy, Curr. Opin. Pharmacol. 8 (2008) 627–631.
antibody ratio of about 3.5 to treat patients with HER2-positive [7] R.M. Lu, Y.C. Hwang, I.J. Liu, C.C. Lee, H.Z. Tsai, H.J. Li, H.C. Wu, Development of
metastatic breast cancer [108,113–116]. After the initial binding therapeutic antibodies for the treatment of diseases, J. Biomed. Sci. 27 (2020)
to the IV sub-domain of HER2 receptor, the ADC undergoes 1–30.
[8] H. Kaplon, M. Muralidharan, Z. Schneider, J.M. Reichert, Antibodies to watch
receptor-mediated internalization and lysosomal degradation.
in 2020, mAbs 12 (2020) 1703531–1703554.
The cytotoxic small molecule released in breast cancer cells binds [9] P. Hubert, S. Amigorena, Antibody-dependent cell cytotoxicity in monoclonal
tubulin, disrupts microtubule synthesis, and ultimately leads to antibody-mediated tumor immunotherapy, Oncoimmunology 1 (2012) 103–
105.
apoptosis and cell death. The clinical data showed that trastuzu-
[10] G. Toth, A. Szoor, L. Simon, Y. Yarden, J. Szollosi, G. Vereb, The combination of
mab emtansine treatment improved the median progression-free trastuzumab and pertuzumab administered at approved doses may delay
survival of 6.2 (95% CI 5.59–6.87) with reduced incidence from development of trastuzumab resistance by additively enhancing antibody-
3.3 months (2.89–4.14) in control group [117]. dependent cell-mediated cytotoxicity, mAbs 8 (2016) 1361–1370.
[11] D. Ernst, B.A. Williams, X.H. Wang, N. Yoon, K.P. Kim, J. Chiu, Z.J. Luo, K.G.
Hermans, J. Krueger, A. Keating, Humanized anti-CD123 antibody facilitates
NK cell antibody-dependent cell-mediated cytotoxicity (ADCC) of Hodgkin
5. Market Value and Technical Trend of mAb-based Therapies lymphoma targets via ARF6/PLD-1, Blood Cancer J. 9 (2019) 6–16.
[12] T. Nakajima, H. Okayama, M. Ashizawa, M. Noda, K. Aoto, M. Saito, T. Monma,
With high therapeutic efficacy, mAbs have emerged as a major S. Ohki, M. Shibata, S. Takenoshita, K. Kono, Augmentation of antibody-
dependent cellular cytotoxicity with defucosylated monoclonal antibodies in
targeted treatment strategy of cancers, where the global market patients with GI-tract cancer, Oncol. Lett. 15 (2018) 2604–2610.
value of therapeutic mAbs was about 150 billion USD in 2019. Con- [13] L.M. Rogers, S. Veeramani, G.J. Weiner, Complement in monoclonal antibody
sidering the numerous mAbs evaluated in clinical trials, the market therapy of cancer, Immunol. Res. 59 (2014) 203–210.
[14] M.T. Winkler, R.T. Bushey, E.B. Gottlin, M.J. Campa, E.S. Guadalupe, A.D.
will continue to grow with an estimated value of 300 billion USD
Volkheimer, J.B. Weinberg, E.F. Patz Jr., Enhanced CDC of B cell chronic
by 2025 [7]. The ADCs-based cancer therapies have made promis- lymphocytic leukemia cells mediated by rituximab combined with a novel
ing progress over the past decade because of the combined thera- anti-complement factor H antibody, PLoS ONE 12 (2017) e0179841–0179852.
peutic values. The development and modification of new drugs, [15] G. Kohler, C. Milstein, Continuous cultures of fused cells secreting antibody of
predefined specificity, Nature 256 (1975) 495–497.
development of novel cancer-targeted mAbs (e.g. humanized [16] M. Tomita, K. Tsumoto, Hybridoma technologies for antibody production,
mAbs), and design of new linkers to improve plasma stability Immunotherapy. 3 (2011) 371–380.
306 Y. Si et al. / Chinese Journal of Chemical Engineering 30 (2021) 301–307

[17] J.G.R. Hurrell, Monoclonal Hybridoma Antibodies: Techniques and [50] H. Liu, K. May, Disulfide bond structures of IgG molecules, mAbs 4 (2012) 17–
Applications, CRC Press, 2018. 23.
[18] S. Zaroff, G. Tan, Hybridoma technology: the preferred method for [51] G. Vidarsson, G. Dekkers, T. Rispens, IgG subclasses and allotypes: from
monoclonal antibody generation for in vivo applications, Biotechniques 67 structure to effector functions, Front. Immunol. 5 (2014) 1–17.
(2019) 90–92. [52] X.-R. Jiang, A. Song, S. Bergelson, T. Arroll, B. Parekh, K. May, S. Chung, R.
[19] P. Holzlohner, K. Hanack, Generation of murine monoclonal antibodies by Strouse, A. Mire-Sluis, M. Schenerman, Advances in the assessment and
hybridoma technology, J. Visualized Exp. (2017) e54832–54838. control of the effector functions of therapeutic antibodies, Nat. Rev. Drug
[20] C. Zhang, Hybridoma technology for the generation of monoclonal antibodies, Discovery 10 (2011) 101–111.
Methods Mol. Biol. 901 (2012) 117–135. [53] Z.K. Indik, J. Park, S. Hunter, A.D. Schreiber, The molecular dissection of Fcy
[21] R.M. Hnasko, L.H. Stanker, Hybridoma technology, Methods Mol. Biol. 1318 receptor mediated phagocytosis, J. Am. Soc. Hematol. 86 (1995) 4389–4399.
(2015) 15–28. [54] L.M. Weiner, R. Surana, S. Wang, Monoclonal antibodies: versatile platforms
[22] S. Pandey, Hybridoma technology for production of monoclonal antibodies, for cancer immunotherapy, Nat. Rev. Immunol. 10 (2010) 317–327.
Hybridoma 1 (2010) 88–94. [55] C. Janeway, Immunobiology: The Immune System in Health and Disease,
[23] R.J. Ober, C.G. Radu, V. Ghetie, E.S. Ward, Differences in promiscuity for Garland Science, Taylor & Francis Group, 1999.
antibody-FcRn interactions across species: implications for therapeutic [56] A.F. LoBuglio, R.H. Wheeler, J. Trang, A. Haynes, K. Rogers, E.B. Harvey, L. Sun,
antibodies, Int. Immunol. 13 (2001) 1551–1559. J. Ghrayeb, M.B. Khazaeli, Mouse/human chimeric monoclonal antibody in
[24] M. Stern, R. Herrmann, Overview of monoclonal antibodies in cancer therapy: man: Kinetics and immune response, Proc. Natl. Acad. Sci. USA 86 (1989)
present and promise, Crit. Rev. Oncol. Hematol. 54 (2005) 11–29. 4220–4224.
[25] C.M. Hammers, J.R. Stanley, Antibody phage display: technique and [57] J.J. Tjandra, L. Ramadi, I.F. McKenzie, Development of human anti-murine
applications, J, Invest. Dermatol. 134 (2014) 1–5. antibody (HAMA) response in patients, Immunol. Cell Biol. 68 (1990) 367–376.
[26] M.A. Alfaleh, H.O. Alsaab, A.B. Mahmoud, A.A. Alkayyal, M.L. Jones, S.M. [58] S.L. Morrison, M.J. Johnson, L.A. Herzenberg, V.T. Oi, Chimeric human
Mahler, A.M. Hashem, Phage display derived monoclonal antibodies: from antibody molecules: mouse antigen-binding domains with human constant
bench to bedside, Front. Immunol. 11 (2020) 1986–2022. region domains, Proc. Natl. Acad. Sci. USA 81 (1984) 6851–6855.
[27] M.L. Chiu, G.L. Gilliland, Engineering antibody therapeutics, Curr. Opin. Struct. [59] L.G. Presta, Engineering of therapeutic antibodies to minimize
Biol. 38 (2016) 163–173. immunogenicity and optimize function, Adv. Drug Deliv. Rev. 58 (2006)
[28] S.A. Sievers, L. Scharf, A.P. West Jr., P.J. Bjorkman, Antibody engineering for 640–656.
increased potency, breadth and half-life, Curr. Opin. HIV AIDS 10 (2015) 151–159. [60] P.T. Jones, P.H. Dear, J. Foote, M.S. Neuberger, G. Winter, Replacing the
[29] A. Thakur, M. Huang, L.G. Lum, Bispecific antibody based therapeutics: complementarity-determining regions in a human antibody with those from
Strengths and challenges, Blood Rev. 32 (2018) 339–347. a mouse, Nature 321 (1986) 522–525.
[30] U. Brinkmann, R.E. Kontermann, The making of bispecific antibodies, mAbs 9 [61] J. McCafferty, A.D. Griffiths, G. Winter, D.J. Chiswell, Phage antibodies:
(2017) 182–212. filamentous phage displaying antibody variable domains, Nature 348 (1990)
[31] Z. Liu, E.C. Leng, K. Gunasekaran, M. Pentony, M. Shen, M. Howard, J. Stoops, K. 552–554.
Manchulenko, V. Razinkov, H. Liu, W. Fanslow, Z. Hu, N. Sun, H. Hasegawa, R. [62] G. Winter, A.D. Griffiths, R.E. Hawkins, H.R. Hoogenboom, Making antibodies
Clark, I.N. Foltz, W. Yan, A novel antibody engineering strategy for making by phage display technology, Annu. Rev. Immunol. 12 (1994) 433–455.
monovalent bispecific heterodimeric IgG antibodies by electrostatic steering [63] H. Thie, T. Meyer, T. Schirrmann, M. Hust, S. Dubel, Phage display derived
mechanism, J. Biol. Chem. 290 (2015) 7535–7562. therapeutic antibodies, Curr. Pharm. Biotechnol. 9 (2008) 439–446.
[32] N. Tsurushita, P.R. Hinton, S. Kumar, Design of humanized antibodies: from [64] G.L. Boulianne, N. Hozumi, M.J. Shulman, Production of functional chimaeric
anti-Tac to Zenapax, Methods 36 (2005) 69–83. mouse/human antibody, Nature 312 (1984) 643–646.
[33] A. Jakobovits, R.G. Amado, X. Yang, L. Roskos, G. Schwab, From XenoMouse [65] N. Lonberg, L.D. Taylor, F.A. Harding, M. Trounstine, K.M. Higgins, S.R.
technology to panitumumab, the first fully human antibody product from Schramm, C.C. Kuo, R. Mashayekh, K. Wymore, J.G. McCabe, et al., Antigen-
transgenic mice, Nat. Biotechnol. 25 (2007) 1134–1143. specific human antibodies from mice comprising four distinct genetic
[34] A. Frenzel, T. Schirrmann, M. Hust, Phage display-derived human antibodies modifications, Nature 368 (1994) 856–859.
in clinical development and therapy, mAbs 8 (2016) 1177–1194. [66] L.L. Green, M.C. Hardy, C.E. Maynard-Currie, H. Tsuda, D.M. Louie, M.J.
[35] P. Tyagi, Recent results and ongoing trials with panitumumab (ABX-EGF), a Mendez, H. Abderrahim, M. Noguchi, D.H. Smith, Y. Zeng, N.E. David, H. Sasai,
fully human anti-epidermal growth factor receptor antibody, in metastatic D. Garza, D.G. Brenner, J.F. Hales, R.P. McGuinness, D.J. Capon, S. Klapholz, A.
colorectal cancer, Clin. Colorectal.Cancer 5 (2005) 21–23. Jakobovits, Antigen-specific human monoclonal antibodies from mice
[36] H. Waldmann, Human monoclonal antibodies: the benefits of humanization, engineered with human Ig heavy and light chain YACs, Nat. Genet. 7 (1994)
Methods Mol. Biol. 2019 (1904) 1–10. 13–21.
[37] L. Loo, M.K. Robinson, G.P. Adams, Antibody engineering principles and [67] M. Brüggemann, M.J. Osborn, B. Ma, J. Hayre, S. Avis, B. Lundstrom, R. Buelow,
applications, Cancer J. 14 (2008) 149–153. Human antibody production in transgenic animals, Archivum immunologiae et
[38] D.G. Maloney, A.J. Grillo-Lopez, C.A. White, D. Bodkin, R.J. Schilder, J.A. therapiae experimentalis 63 (2015) 101–108.
Neidhart, N. Janakiraman, K.A. Foon, T.M. Liles, B.K. Dallaire, K. Wey, I. [68] N. Lonberg, Fully human antibodies from transgenic mouse and phage display
Royston, T. Davis, R. Levy, IDEC-C2B8 (Rituximab) anti-CD20 monoclonal platforms, Curr. Opin. Immunol. 20 (2008) 450–459.
antibody therapy in patients with relapsed low-grade non-Hodgkin’s [69] G. Galizia, E. Lieto, F. De Vita, M. Orditura, P. Castellano, T. Troiani, V.
lymphoma, Blood 90 (1997) 2188–2195. Imperatore, F. Ciardiello, Cetuximab, a chimeric human mouse anti-
[39] D.G. Maloney, A.J. Grillo-Lopez, D.J. Bodkin, C.A. White, T.M. Liles, I. Royston, epidermal growth factor receptor monoclonal antibody, in the treatment of
C. Varns, J. Rosenberg, R. Levy, IDEC-C2B8: results of a phase I multiple-dose human colorectal cancer, Oncogene 26 (2007) 3654–3660.
trial in patients with relapsed non-Hodgkin’s lymphoma, J. Clin. Oncol. Off. J. [70] A.Y. Liu, R.R. Robinson, K.E. Hellstron, E.D. Murray, C.P. Chang, I. Hellstrom,
Am. Soc. Clin. Oncol. 15 (1997) 3266–3274. Chimeric mouse-human IgG1 antibody that can mediate lysis of cancer cells,
[40] D. Zahavi, L. Weiner, Monoclonal antibodies in cancer therapy, Antibodies 9 Proc. Natl. Acad. Sci. USA 84 (1987) 3439–3443.
(2020) 34–53. [71] N.I. Goldstein, M. Prewett, K. Zuklys, P. Rockwell, J. Mendelsohn, Biological
[41] J.K.H. Liu, The history of monoclonal antibody development – Progress, efficacy of a chimeric antibody to the epidermal growth factor receptor in a
remaining challenges and future innovations, Ann. Med. Surg. 3 (2014) 113– human Tumor Xenograft model, Clin. Cancer Res. 1 (1995) 1311–1318.
116. [72] J. Mendelsohn, M. Prewett, P. Rockwell, N.I. Goldstein, CCR 20th anniversary
[42] N. Stergiou, J. Nagel, S. Pektor, A.S. Heimes, J. Jakel, W. Brenner, M. Schmidt, commentary: a chimeric antibody, C225, inhibits EGFR activation and tumor
M. Miederer, H. Kunz, F. Roesch, E. Schmitt, Evaluation of a novel monoclonal growth, Clin. Cancer Res. 21 (2015) 227–229.
antibody against tumor-associated MUC1 for diagnosis and prognosis of [73] M. Changes, Highlights of Prescribing Information-cetuximab, Package Insert
breast cancer, Int. J. Med. Sci. 16 (2019) 1188–1198. 50 (2004) 1–25. http://pi.lilly.com/us/zyprexa-pi.pdf.
[43] X. Zhang, G. Soori, T.J. Dobleman, G.G. Xiao, The application of monoclonal [74] S. Maximiano, P. Magalhaes, M.P. Guerreiro, M. Morgado, Trastuzumab in the
antibodies in cancer diagnosis, Expert Rev. Mol. Diagn. 14 (2014) 97–106. treatment of breast cancer, BioDrugs 30 (2016) 75–86.
[44] R.P. Taylor, M.A. Lindorfer, The role of complement in mAb-based therapies of [75] A. Hajjar, M.A. Ergun, O. Alagoz, M. Rampurwala, Cost-effectiveness of
cancer, Methods 65 (2014) 18–27. adjuvant paclitaxel and trastuzumab for early-stage node-negative, HER2-
[45] K.J. Hamblett, P.D. Senter, D.F. Chace, M.M. Sun, J. Lenox, C.G. Cerveny, K.M. positive breast cancer, PLoS ONE 14 (2019) e0217778–0217791.
Kissler, S.X. Bernhardt, A.K. Kopcha, R.F. Zabinski, D.L. Meyer, J.A. Francisco, [76] S.M. Tolaney, W.T. Barry, C.T. Dang, D.A. Yardley, B. Moy, P.K. Marcom, K.S.
Effects of drug loading on the antitumor activity of a monoclonal antibody Albain, H.S. Rugo, M. Ellis, I. Shapira, A.C. Wolff, L.A. Carey, B.A. Overmoyer, A.
drug conjugate, Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 10 (2004) 7063– H. Partridge, H. Guo, C.A. Hudis, I.E. Krop, H.J. Burstein, E.P. Winer, Adjuvant
7070. paclitaxel and trastuzumab for node-negative, HER2-positive breast cancer,
[46] A. Wolska-Washer, T. Robak, Safety and tolerability of antibody-drug New Engl. J. Med. 372 (2015) 134–141.
conjugates in cancer, Drug Saf. 42 (2019) 295–314. [77] M.A. Molina, J. Codony-Servat, J. Albanell, F. Rojo, J. Arribas, J. Baselga,
[47] Y. Si, S. Kim, E. Zhang, Y. Tang, R. Jaskula-Sztul, J.M. Markert, H. Chen, L. Zhou, Trastuzumab (Herceptin), a Humanized Anti-HER2 Receptor Monoclonal
X.M. Liu, Targeted exosomes for drug delivery: biomanufacturing, surface Antibody, Inhibits Basal and Activated HER2 Ectodomain Cleavage in Breast
tagging, and validation, Biotechnol. J. 15 (2020) e1900163–1900174. Cancer Cells, Cancer Research 61 (2001) 4744–4749.
[48] W. Wang, S. Singh, D.L. Zeng, K. King, S. Nema, Antibody Structure, Instability, [78] L. Jiang, T. Jiang, J. Luo, Y. Kang, Y. Tong, X. Song, X. Gao, W. Yao, H. Tian,
and Formulation, J. Pharm. Sci. 96 (2007) 1–26. Efficient acquisition of fully human antibody genes against self-proteins by
[49] R. Jefferis, J. Lund, Interaction sites on human IgG-Fc for FccR: current models, sorting single B cells stimulated with vaccines based on nitrated T helper cell
Immunol. Lett. 82 (2002) 57–65. epitopes, J. Immunol. Res. 2019 (2019) 7914326–7914341.
Y. Si et al. / Chinese Journal of Chemical Engineering 30 (2021) 301–307 307

[79] I. Hellmann, L. Waldmeier, M.C. Bannwarth-Escher, K. Maslova, F.I. Wolter, U. [101] M. Gottardi, F. Mosna, S. de Angeli, C. Papayannidis, A. Candoni, M. Clavio, C.
Grawunder, R.R. Beerli, Novel antibody drug conjugates targeting tumor- Tecchio, A. Piccin, M.C. dell’Orto, F. Benedetti, G. Martinelli, F. Gherlinzoni,
associated receptor tyrosine kinase ROR2 by functional screening of fully Clinical and experimental efficacy of gemtuzumab ozogamicin in core
human antibody libraries using transpo-mAb display on progenitor B cells, binding factor acute myeloid leukemia, Hematol Rep. 9 (2017) 87–90.
Front. Immunol. 9 (2018) 2490–2505. [102] F.R. Appelbaum, I.D. Bernstein, Gemtuzumab ozogamicin for acute myeloid
[80] B. Boll, H. Hansen, F. Heuck, K. Reiners, P. Borchmann, A. Rothe, A. Engert, E. leukemia, Blood 130 (2017) 2373–2376.
Pogge von Strandmann, The fully human anti-CD30 antibody 5F11 activates [103] J.K. Lamba, L. Chauhan, M. Shin, M.R. Loken, J.A. Pollard, Y.C. Wang, R.E. Ries,
NF-{kappa}B and sensitizes lymphoma cells to bortezomib-induced R. Aplenc, B.A. Hirsch, S.C. Raimondi, R.B. Walter, I.D. Bernstein, A.S. Gamis, T.
apoptosis, Blood 106 (2005) 1839–1842. A. Alonzo, S. Meshinchi, CD33 Splicing polymorphism determines
[81] A.L. Nelson, E. Dhimolea, J.M. Reichert, Development trends for human gemtuzumab ozogamicin response in De Novo Acute Myeloid Leukemia:
monoclonal antibody therapeutics, Nat. Rev. Drug Discov. 9 (2010) 767–774. report from randomized phase III children’s oncology group trial AAML0531,
[82] Q. Wang, Y. Chen, J. Park, X. Liu, Y. Hu, T. Wang, K. McFarland, M.J. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 35 (2017) 2674–2682.
Betenbaugh, Design and production of bispecific antibodies, Antibodies (Basel) [104] C.D. Godwin, R.P. Gale, R.B. Walter, Gemtuzumab ozogamicin in acute
8 (2019), antib8030043-8030062. myeloid leukemia, Leukemia 31 (2017) 1855–1868.
[83] J. Golay, S. Choblet, J. Iwaszkiewicz, P. Cerutti, A. Ozil, S. Loisel, M. Pugniere, G. [105] C. Selby, L.R. Yacko, A.E. Glode, Gemtuzumab ozogamicin: back again, J. Adv.
Ubiali, V. Zoete, O. Michielin, C. Berthou, J. Kadouche, J.P. Mach, M. Duonor- Pract. Oncol. 10 (2019) 68–82.
Cerutti, Design and validation of a novel generic platform for the production [106] J. Baron, E.S. Wang, Gemtuzumab ozogamicin for the treatment of acute
of tetravalent IgG1-like bispecific antibodies, J. Immunol. 196 (2016) 3199– myeloid leukemia, Expert Rev. Clin. Pharmacol. 11 (2018) 549–559.
3211. [107] S. Castaigne, C. Pautas, C. Terré, E. Raffoux, D. Bordessoule, J.-N. Bastie, O.
[84] F. Liu, S. Guttikonda, M.R. Suresh, Bispecific monoclonal antibodies against a Legrand, X. Thomas, P. Turlure, O. Reman, T. de Revel, L. Gastaud, N. de
viral and an enzyme: utilities in ultrasensitive virus ELISA and phage display Gunzburg, N. Contentin, E. Henry, et al., Effect of gemtuzumab ozogamicin on
technology, J. Immunol. Methods 274 (2003) 115–127. survival of adult patients with de-novo acute myeloid leukaemia (ALFA-0701):
[85] S.E. Sedykh, V.V. Prinz, V.N. Buneva, G.A. Nevinsky, Bispecific antibodies: a randomised, open-label, phase 3 study, Lancet 379 (2012) 1508–1516.
design, therapy, perspectives, Drug Des. Devel. Ther. 12 (2018) 195–208. [108] D.S. Pereira, C.I. Guevara, L. Jin, N. Mbong, A. Verlinsky, S.J. Hsu, H. Avina, S.
[86] K.E. Tiller, P.M. Tessier, Advances in antibody design, Annu. Rev. Biomed. Eng. Karki, J.D. Abad, P. Yang, S.J. Moon, F. Malik, M.Y. Choi, Z. An, K. Morrison,
17 (2015) 191–216. et al., AGS67E, an anti-CD37 monomethyl auristatin E antibody-drug
[87] Z. Elgundi, M. Reslan, E. Cruz, V. Sifniotis, V. Kayser, The state-of-play and conjugate as a potential therapeutic for B/T-cell malignancies and AML: a
future of antibody therapeutics, Adv. Drug Deliv. Rev. 122 (2017) 2–19. new role for CD37 in AML, Mol. Cancer Ther. 14 (2015) 1650–1660.
[88] M. Godar, H. de Haard, C. Blanchetot, J. Rasser, Therapeutic bispecific [109] N.C. Richardson, Y.L. Kasamon, H. Chen, R.A. de Claro, J. Ye, G.M. Blumenthal,
antibody formats: a patent applications review (1994–2017), Expert Opin. A.T. Farrell, R. Pazdur, FDA approval summary: brentuximab vedotin in first-
Ther. Pat. 28 (2018) 251–276. line treatment of peripheral T-cell lymphoma, Oncologist 24 (2019) e180–
[89] L.L. Sun, D. Ellerman, M. Mathieu, M. Hristopoulos, X. Chen, Y. Li, X. Yan, R. e187.
Clark, A. Reyes, E. Stefanich, E. Mai, J. Young, C. Johnson, M. Huseni, X. Wang, [110] S. Horwitz, O.A. O’Connor, B. Pro, T. Illidge, M. Fanale, R. Advani, N.L. Bartlett,
et al., Anti-CD20/CD3 T cell-dependent bispecific antibody for the treatment J.H. Christensen, F. Morschhauser, E. Domingo-Domenech, G. Rossi, W.S. Kim,
of B cell malignancies, Sci. Transl. Med. 7 (2015), 287ra270-279. T. Feldman, A. Lennard, D. Belada, et al., Brentuximab vedotin with
[90] H. Yao, F. Jiang, A. Lu, G. Zhang, Methods to design and synthesize antibody- chemotherapy for CD30-positive peripheral T-cell lymphoma (ECHELON-2):
drug conjugates (ADCs), Int. J. Mol. Sci. 17 (2016) 194–205. a global, double-blind, randomised, phase 3 trial, Lancet 393 (2019) 229–240.
[91] J.R. McCombs, S.C. Owen, Antibody drug conjugates: design and selection of [111] C. van der Weyden, M. Dickinson, J. Whisstock, H.M. Prince, Brentuximab
linker, payload and conjugation chemistry, AAPS J. 17 (2015) 339–351. vedotin in T-cell lymphoma, Expert Rev. Hematol. 12 (2019) 5–19.
[92] L.R. Saunders, A.J. Bankovich, W.C. Anderson, M.A. Aujay, S. Bheddah, K. Black, [112] H.M. Prince, Y.H. Kim, S.M. Horwitz, R. Dummer, J. Scarisbrick, P. Quaglino, P.
R. Desai, P.A. Escarpe, J. Hampl, A. Laysang, D. Liu, J. Lopez-Molina, M. Milton, L. Zinzani, P. Wolter, J.A. Sanches, P.L. Ortiz-Romero, O.E. Akilov, L. Geskin, J.
A. Park, M.A. Pysz, et al., A DLL3-targeted antibody-drug conjugate eradicates Trotman, K. Taylor, S. Dalle, et al., Brentuximab vedotin or physician’s choice
high-grade pulmonary neuroendocrine tumor-initiating cells in vivo, Sci. in CD30-positive cutaneous T-cell lymphoma (ALCANZA): an international,
Transl. Med. 7 (2015), 302ra136-163. open-label, randomised, phase 3, multicentre trial, Lancet 390 (2017) 555–
[93] L. Zhou, N. Xu, Y. Sun, X.M. Liu, Targeted biopharmaceuticals for cancer 566.
treatment, Cancer Lett. 352 (2014) 145–151. [113] Z. Zolcsak, D. Loirat, A. Fourquet, Y.M. Kirova, Adjuvant Trastuzumab
[94] H. Almasbak, T. Aarvak, M.C. Vemuri, CAR T cell therapy: a game changer in Emtansine (T-DM1) and concurrent radiotherapy for residual invasive
cancer treatment, J. Immunol. Res. 2016 (2016) 5474602–5474611. HER2-positive breast cancer: single-center preliminary results, Am. J. Clin.
[95] H. Dai, Y. Wang, X. Lu, W. Han, Chimeric antigen receptors modified T-cells Oncol. 43 (12) (2020) 895–901.
for cancer therapy, J. Natl Cancer Inst. 108 (2016), djv439-552. [114] K.A. Lyseng-Williamson, Trastuzumab emtansine: a review of its adjuvant
[96] M.S. Magee, A.E. Snook, Challenges to chimeric antigen receptor (CAR)-T cell use in residual invasive HER2-positive early breast cancer, Drugs 80 (2020)
therapy for cancer, Discov. Med. 18 (2014) 265–271. 1723–1730.
[97] B.L. Zhang, D.Y. Qin, Z.M. Mo, Y. Li, W. Wei, Y.S. Wang, W. Wang, Y.Q. Wei, [115] G. von Minckwitz, C.S. Huang, M.S. Mano, S. Loibl, E.P. Mamounas, M. Untch,
Hurdles of CAR-T cell-based cancer immunotherapy directed against solid N. Wolmark, P. Rastogi, A. Schneeweiss, A. Redondo, H.H. Fischer, W. Jacot, A.
tumors, Sci. China Life Sci. 59 (2016) 340–348. K. Conlin, C. Arce-Salinas, I.L. Wapnir, et al., Trastuzumab emtansine for
[98] R. Kunert, D. Reinhart, Advances in recombinant antibody manufacturing, residual invasive HER2-positive breast cancer, New Engl. J. Med. 380 (2019)
Appl. Microbiol. Biotechnol. 100 (2016) 3451–3461. 617–628.
[99] P. Polakis, Antibody drug conjugates for cancer therapy, Pharmacol. Rev. 68 [116] M. Barok, H. Joensuu, J. Isola, Trastuzumab emtansine: mechanisms of action
(2016) 3–19. and drug resistance, Breast Cancer Res. 16 (2014) 209–221.
[100] C.C. Zhang, Z. Yan, B. Pascual, A. Jackson-Fisher, D.S. Huang, Q. Zong, M. Elliott, [117] I.E. Krop, S.-B. Kim, A. González-Martín, P.M. LoRusso, J.-M. Ferrero, M. Smitt,
C. Fan, N. Huser, J. Lee, M. Sung, P. Sapra, Gemtuzumab Ozogamicin (GO) R. Yu, A.C.F. Leung, H. Wildiers, Trastuzumab emtansine versus treatment of
inclusion to induction chemotherapy eliminates leukemic initiating cells and physician’s choice for pretreated HER2-positive advanced breast cancer
significantly improves survival in mouse models of acute myeloid Leukemia, (TH3RESA): a randomised, open-label, phase 3 trial, Lancet Oncol. 15 (2014)
Neoplasia 20 (2018) 1–11. 689–699.

You might also like