The Wonderful Story of Monoclonal Antibodies: Marie C Béné
The Wonderful Story of Monoclonal Antibodies: Marie C Béné
The Wonderful Story of Monoclonal Antibodies: Marie C Béné
DOI: 10.1111/ijlh.13006
SUPPLEMENT ARTICLE
Marie C Béné
KEYWORDS
biologics, flow cytometry, immunoglobulins, monoclonal antibodies, pathology
8 | wileyonlinelibrary.com/journal/ijlh
© 2019 John Wiley & Sons Ltd Int J Lab Hematol. 2019;41(Suppl. 1):8–14.
BÉNÉ |
9
TA B L E 1 Proposed terminology for immunoglobulin fragments, adapted from the nomenclature of human immunoglobulins, Immunology
1963
horses and rabbits have to be commended for having produced Biochemical studies, using mostly crystallography and sequenc-
protective sera, rich in specific immunoglobulins, that, among ing, meanwhile tried to better understand the structure of these gly-
other feats, saved soldiers of World wars from tetanus, diphtheria coproteins. The striking juxtaposition of molecular motifs of about
or other ailments. 100 amino acids including cysteines strategically placed to allow
The era of serotherapy blossomed and progressed. Many re- the formation of disulfide bonds was progressively identified as an
12
searchers, Edwin Cohn among others, devised ways to purify extremely clever structure. Further modelization highlighted the
human immunoglobulins in order to provide patients that needed systematic organization of these amino acids in antiparallel beta-
it with passive immunization. This led to precious preparations di- pleated sheets linked by loose loops. The notion of domain emerged.
rected toward infectious agents, helpful to alleviate infections in The fact that this molecular structure allowed for adhesion between
immunocompromised/immunodeficient patients. Broader prepara- siblings and was in fact used by quite a number of transmembrane
tions of IVIG or intravenous immunoglobulins were developed, still or extracellular glycoproteins progressively led to the notion of “im-
widely used not only for immunodeficient patients but also for the munoglobulin superfamily”. 20 Indeed, the unique assembly of these
treatment of such autoimmune disorders as immune thrombocyto- domains confers them with a striking capacity to interact together.
13
penic purpura. Nowadays, IVIG are processed in order to try and However, even though these notions became increasingly clear,
limit the adverse reactions they may yield.14 the exquisite specificity of immunoglobulins for a given antigen, al-
though clearly carried by their variable domains, remained a mystery.
2 | D I S COV E RY O F A N A M A Z I N G
M O LEC U L A R S TRU C T U R E 3 | R I S E O F M O N O C LO N A L A NTI B O D I E S
But much work and time were still needed to unravel the mysteries In the 1970s, this question of variability was gnawing at the brilliant
of immunoglobulins. This was helped by refining animal immuniza- mind of Cesar Milstein, a researcher of Argentinian origin, work-
tion techniques but also by the characterization of myelomatous ing in London in the renowned laboratory of Frederick Sanger. The
proteins from patients with multiple myeloma. By the time the WHO model developed by Cesar Milstein was that of rabbits immunized
nomenclature was published in the early 60s,8 the basic structure with sheep red blood cells. His tools were antibody purification,
of immunoglobulins had been deciphered, a model of the associa- crystallography, then RNA sequencing, but he was puzzled by the
tion of heavy and light chains having been proposed in 1962 by differences observed each time he was using a new batch of rab-
Rodney Porter.15 Heavy chain isotypes had been described for IgG bit antiserum. Because he believed that somatic mutations were
and IgM in the 1940s, IgA in 1956, and IgD in 1964. IgE would have responsible for the generation of specific immunoglobulins, he em-
to wait until 1967 to be characterized and linked to the long-known barked on long-term culture of cell lines or myeloma cells. He thus
“reagins.” The light chains of immunoglobulins had been initially de- participated to experiments conducted on hybridomas generated
scribed as proteins present in the urine of myelomatous patients by with Abraham Karpas and Dick Cotton, following observations by
Henry Bence Jones as early as 1864.16 That there were two types of Hervé Bazin in Belgium. Immortalization of a rat myeloma with a
Bence Jones proteins was however only elucidated almost a century mouse myeloma after fusion by the Sendai virus resulted in a hybri-
later, in 1950 by Korngold and Lipari who used the first letter of their doma producing both types of myelomatous immunoglobulins. After
respective names to give them their Greek equivalent denomination identification of the two respective RNAs in the cells, the conclusion
of kappa and lambda chains.17 Further chemical and enzymatic ex- was that each had brought, from its nucleus, the genetic message of
periments allowed to discover that the four-chain structure could each immunoglobulin. The big problem was that these immunoglob-
be subdivided in fragments of different functions (Table 1), their ulins had no known reactivity toward a recognized antigen. After a
nomenclature being also clarified in 19648 mostly as Fc (for crystal- talk he gave in the Basel Institute, he was approached by a young fel-
lizable) and Fab (for antigen-binding sometimes also referred to as low named George Kohler, fascinated by the topic and who applied
antibody fragment). to come as a postdoctoral fellow in Cambridge. There, as an ancil-
In the 1960s, several seminal works also first identified that lym- lary project, Milstein asked him if he could devise a way to obtain a
phocytes were the cells responsible for specific immunity,18 then steadily similar batch of anti-sheep red blood cell immunoglobulins.
that the production of antibodies was the realm of B cells.19 Aware of the hybridoma experiment, Kohler grew the idea to fusion
|
10 BÉNÉ
not two myelomas but B cells from an immunized animal (with sheep were published in a mythic book in 1984. 22 Thanks to the statisti-
red blood cells) and an immortal myeloma. The screen for hybrido- cal gifts of Catherine Hill at Gustave Roussy cancer center, results
mas producing one single species of anti-sheep red blood cells im- from 55 research groups were computed to group within clusters the
munoglobulins was successful. The cells were growing as a single 139 antibodies provided by different laboratories. All antibodies in a
clone, indefinitely, and secreted unlimited amounts of the immuno- cluster displayed the same reactivity. This statistical approach is the
globulin encoded by their DNA: monoclonal antibodies! Milstein was one that yielded the name “Cluster of differentiation” or CD. Because
first thrilled to have a tool allowing to test (unsuccessfully) his theory so many of the new “antigens” discovered were utterly unknown,
of somatic mutations. However, the two scientists soon recognized they were attributed the same name. CD is thus a denomination
that they had something much bigger, allowing to produce “à la carte” characterizing monoclonal antibodies of the same specificity or the
antibodies. molecule thus identified. The first HLDA confirmed 11 specificities
and proposed four additional provisional ones, numbered from CD1
to CDw15. After the 10th, in 2014 in Barcelona, 371 CD have been
4 | M O N O C LO N A L A NTI B O D I E S A S N E W formally recognized, quite a number of them being subdivided in
R E AG E NT S subcluster isoforms. 26 Upon regular international meetings, the rules
were refined, requiring at least two antibodies from two different
Kohler and Milstein, after a rebuttal in 1975 from the journal Science, labs to have been reported, together with the protein and later gene
finally published their outbreaking results in a short letter in Nature structure of the related molecule. The production of these reagents
21
in August of the same year. This initiated a revolution and much left mice peritoneum to reach the more ethical fermenters allow-
ebullition in the immunological world. The technique by itself was ing for mass production. Along the way, however, some extremely
rather simple. It involved immunization of mice with small doses of precious hybridomas were lost and some were never recognized for
antigen, removal of their spleen, and mixture of splenic B-cells with their possible value. Indeed, we still are far away from of the ex-
a rat myeloma cell line. Sendai virus, then polyethylene glycol, were pected 7000 cell proteins that could be identified by such tools. 27 Of
found to achieve the random hybridization of cells. Use of a selective note, some monoclonal antibodies have also largely been produced
medium allowed only hybridomas of a B-lymphocyte and a myeloma toward already known structures of which they did not change the
cell to survive. The trickiest part, probably, was the tedious seeding name. They, however, provided exquisite tools to study them.
of limited dilutions of the hybridomas, drop by drop in multiple-well Monoclonal antibodies were first used as ascites fluids as men-
plates, with the aim of having no more than one cell in each well. tioned above. The monospecificity and huge affinity of the clonal
After selecting wells seeded with hybridomas and letting the lat- immunoglobulins allowed for extensive research, based on the high
ter grow, another tedious task was to seek for the supernatant(s) level of dilution permitted for experiments in many types of settings,
containing immunoglobulins of the expected specificity. Afterwards, from immunochemistry and immunophenotyping to immunohistol-
mice came back in the picture in the now abandoned peritoneal in- ogy and basic research. The high specificity of monoclonals found its
jection of the selected hybridoma, followed by regular aspiration of way in a number of laboratory applications. Many of the immunore-
the resulting immunoglobulin-rich ascites produced. agents already used in nephelemetry, turbidimetry, ELISA or multi-
The spread was what would now be qualified as “viral.” Hundreds plex chemiluminescence were successfully replaced by monoclonals.
of abstracts and papers blossomed. In the early 1980s, Laurence Even in more fundamental work, Western blots found an increased
Boumsell and Alain Bernard, who had close relationships both with resolution by the use of monoclonals. The dissection of antigenic
Cesar Milstein and Jean Dausset, devised a way to bring some order epitopes became feasible.
to the ongoing folly. Rich of the experience of HLA workshops In the initial applications of immunophenotyping or immu-
heralded by their mentor Dausset, they organized a worldwide nochemistry, indirect methods were used, with anti-mouse labeled
co-operation under the name of Human Leukocyte Differentiation antisera as secondary layer. The reagent industry soon grabbed
Antigen (HLDA) workshops. 22 Indeed, most of the laboratories that the interest of providing purified monoclonal antibody solutions. It
had been investing in this new wave had been trying to use these must be acknowledged that the Ortho company was pioneer in this
new fantastic tools to find a better definition of white blood cells. It field. 28 Very early in the 1980s the antibodies produced by Patrice
must be reminded that, at that time, morphology, some enzymatic Kung were exploited and commercialized by this manufacturer as
reactions, and the use of polyclonal animal antisera were the only “OKT” or “Ortho Kung T”-antibodies. OKT3, OKT4, and OKT8 were
means to (poorly) decipher leukocyte subsets. It was possible to certainly the first used in many laboratories to discover the exquisite
identify immunoglobulins on the surface of mature B cells and in the homeostasis of peripheral T-cell subsets, initially only suspected by
cytoplasm of preB cells. 23,24 T-lymphocytes, characterized after the the use of the abovementioned sophisticated rosette experiments.
work of John Miller on the thymus, 25 were tackled by sophisticated Quickly, fluorochrome-
conjugated monoclonals became available
variations of their ability to collect sheep red blood cells around and greatly helped the development of flow cytometry.
them, the rosetting phenomenon. In this specific field of flow cytometry, the first steps were to
Human Leukocyte Differentiation Antigen workshops met with a move from indirect immunofluorescence toward direct labeling. The
tremendous success right from the start when the first proceedings all-time winner, already developed in the late 70s for fluorescence
BÉNÉ |
11
F I G U R E 1 Nomenclature of therapeutic monoclonal antibodies. “Momabs” are completely murine (and highly immunogenic).
Bioengineered antibodies have been devised to limit murine sequences to the antigen-binding regions. Their various names thus depend
on the variable region of the immunoglobulin. The latter is fully murine in “ximabs”. The difference between humanized (-zu) and fully
human monoclonals (-u) depends on how close to a human sequence is the antigen-binding structure. To produce fully human monoclonals,
the mouse genome is modified to harbor the human immunoglobulin locus before immunization 51,52 [Colour figure can be viewed at
wileyonlinelibrary.com]
TA B L E 2 Examples of different types of therapeutic antibodies for the treatment of hematological malignancies, with decreasing
antigenicity. The isotype used is important for the half-life of the product and to favor or alleviate interactions with Fc receptors
deciphering of antibody-coding regions allowed for a variety of have been devised to modulate these interactions and impact on the
developments to improve the specificity and affinity of therapeu- drug's pharmacokinetics.39 Another strategy is to generate hybrid
tic recombinant monoclonal antibodies. Such strategies also were IgG2-CH1 (which does not bind complement) and IgG4-CH3-CG3
applied to improve the pharmacokinetics of these new drugs, for (which do not bind FcR), preferred for antagonist or agonist anti-
instance favoring their recycling by interaction with the neonatal bodies that should not lead to cytotoxicity. This was applied to the
Fcn receptor. 38 generation of eculizumab, the anti-C5 antibody stabilizing patients
Currently, most of therapeutic monoclonals are thus human with paroxysmal nocturnal hemoglobinuria.40 Indeed, besides cy-
IgG1,34 which recognize the chosen target by their Fab portion totoxicity to remove tumor cells or adverse immune reactions, re-
and display effector functions through their Fc. The latter include cent remarkable applications involve manipulation of the immune
complement binding in complement depending cytotoxicity (CDC) system. The development of immune checkpoint inhibitors reviving
and attachment to Fc receptors (FcR) on phagocytes in antibody- the activity of anergic or exhausted T cells has led to a whole field
dependent cellular phagocytosis (ADCP) and on natural killer cells of antitumoral strategies based on reactivation of the patient's own
in antibody-
dependent cellular cytotoxicity (ADCC). These func- defense systems.41
tions are mostly used for therapeutic preparations directed to Other approaches removed the Fc fragment when the latter was
tumor cells such as lymphoma cells. Variations in Fc glycosylation not necessary. In 1994, abciximab, a chimeric anti-GPIIb/IIIa Fab
F I G U R E 2 A brief history of
immunoglobulins and monoclonal
antibodies [Colour figure can be viewed at
wileyonlinelibrary.com]
BÉNÉ |
13
development, namely bispecific antibodies. In this type of construc- 1. von Behring E, Kitasato S. Über das Zustandekommen der
tion, two variable domains of different specificity are combined in Diphtherie- Immunität und der Tetanus- Immunität bei Thieren.
Dtsch Med Wschr. 1890;16:1113‐1114.
order to connect two different types of cells. The most successful to
2. Lindenmann J. Origin of the terms ‘antibody’ and ‘antigen’. Scand J
date bring T cells, through their anti-CD3 moiety, toward malignant Immunol. 1984;19:281‐285.
B cells expressing CD19.49 More constructions are in progress for 3. Nicolle M. Une conception générale des anticorps et de leurs effets.
other onco-hematologic diseases or other cancers. CR Soc Biol (Paris). 1907;63:77.
4. Deutsch L. Contribution à I’étude de l'origine des anticorps typh-
Finally, following a long series of bioengineering strategies, the
iques. Ann Inst Pasteur. 1899;13:689.
unique ability of immunoglobulins Fab to recognize antigens directly
5. Tizzoni G, Cattani G. Fernere Untersuchungen liber das tetanus-
has been put to use to improve T-cell targeting and activation in fully antitoxin. Zentralbl Bakteriol Mikrobiol Hyg [A]. 1891;10:33.
bioengineered Chimeric Antigen Receptor T cells or CAR T cells. 50 6. Tiselius A. Electrophoresis of serum globulin I. Biochem J.
The Fab portion of an immunoglobulin is modified to produce a sin- 1937;31:313-317.
7. Edelman GM. Dissociation of y- globulin. J Am chem Soc.
gle chain variable fragment (scFv) associating the variable domains
1959;81:3155.
of the heavy and light chain. Genes coding for this construction are 8. Ceppellini R, Dray S, Edelman G, et al. Nomenclature for human
then associated to those encoding a transmembrane portion and in- immunoglobulins. Immunochemistry. 1964;1:145-149.
tracytoplasmic T-cell activation domains. The first CAR T-cells only 9. Heremans JF. Les Globulines Sériques du Système Gamma. Brussels:
Editions Arscia; 1960.
associated the CD3 zeta chain of the T-cell receptor. Nowadays, CAR
10. Ehrlich P. Die Seitenkettentheorie und ihre Gegner. Münchner Med
T cells of three different generations have been produced, associat- Wochenschr 1901;2123‐2124.
ing cytoplasmic portions of other co-activation molecules such as 11. Valent P, Groner B, Schumacher U, et al. Paul Ehrlich (1854-1915)
CD28 and/or 4-1BB. This expanding field triggers increasingly imag- and His Contributions to the Foundation and Birth of Translational
Medicine. J Innate Immun. 2016;8:111-120.
inative solutions, based on the combination of the now well-known
12. Cohn EJ, Oncley JL, Strong LE, Hughes WL Jr, Armstrong SH Jr.
molecular portions of immunoglobulins, T-cell receptors and co- Chemical, clinical, and immunological studies on the products of
activation signals in a genial genetic meccano. Modified T cells from human plasma fractionation. I. The characterization of the protein
the patient are currently used, reinfused after a few days or weeks. fractions of human plasma. J Clin Invest. 1944;23:417‐432.
13. Imbach P, Barandun S, Baumgartner C, Hirt A, Hofer F, Wagner HP.
“Off the shelf” allogeneic CAR T-cells from donors, modified not only
High-dose intravenous gammaglobulin therapy of refractory, in
for their CAR portion but also to remove possible alloreactivity are particular idiopathic, thrombocytopenia in childhood. Helv Paediatr
also developed. Some models even include suicide genes allowing to Acta. 1981;46:81-86.
induce the destruction of the infused CAR T-cells, should they yield 14. Hooper JA. The history and evolution of immunoglobulin products
and their clinical indications. LymphoSign J. 2015;2:181-194.
adverse reactions.
|
14 BÉNÉ
15. Porter RR. The structure of y-globulin and antibodies. In: Gellhorn 35. Trill JJ, Shatzman AR, Ganguly S. Production of monoclonal antibod-
A, Hirschberg E, ed. Basic Problems of Neoplastic Disease. New York, ies in COS and CHO cells. Curr Opin Biotechnol. 1995;6(553):560.
NY: Columbia University Press;1962:177. 36. Sgro C. Side-effects of a monoclonal antibody, muromonab CD3/or-
16. Jones HB. Papers on chemical pathology; prefaced by the thoclone OKT3: a bibliographic review. Toxicology. 1995;105:23e9.
Gulstonian lectures read at the Royal College of Physicians. Lecture 37. Marcus R, Hagenbeek A. The therapeutic use of rituximab in non-
3. Lancet. 1846;2: 88‐92. Hodgkin's lymphoma. Eur J Haematol Suppl. 2007;67(5):14.
17. Korngold L, Lipari R. Multiple myeloma proteins. III. The antigenic 38. Sheridan D, Yu ZX, Zhang Y, et al. Design and preclinical charac-
relationship of Bence-Jones proteins to normal y-globulin and mul- terization of ALXN1210: a novel anti-C5 antibody with extended
tiple myeloma serum proteins. Cancer. 1956;9:262. duration of action. PLoS ONE. 2018;13:e0195909.
18. Gowans JL, McGregor DD, Cowan DM, Ford CE. Initiation of Immune 39. Jefferis R. Posttranslational modifications and the immunogenicity
Responses by Small Lymphocytes. Nature. 1962;196:651-652. of biotherapeutics. J Immunol Res. 2016;2016:5358272.
19. Jerne NK, Henry C, Nordin AA, Fuji H, Koros AM, Lefkovits I. 4 0. Rother RP, Rollins SA, Mojcik CF, Brodsky RA, Bell L. Discovery
Plaque forming cells: methodology and theory. Transplant Rev. and development of the complement inhibitor eculizumab for the
1974;18:130-191. treatment of paroxysmal nocturnal hemoglobinuria. Nat Biotechnol.
20. Halaby DM, Mornon JP. The immunoglobulin superfamily: an in- 2007;25(1256):1264.
sight on its tissular, species, and functional diversity. J Mol Evol. 41. Rotte A, Jin JY, Lemaire V. Mechanistic overview of immune check-
1998;46:389-400. points to support the rational design of their combinations in cancer
21. Köhler G, Milstein C. Continuous cultures of fused cells secreting immunotherapy. Ann Oncol. 2018;29(71):83.
antibody of predefined specificity. Nature. 1975;256:495-497. 42. Foster RH, Wiseman LR. Abciximab. An updated review of its use in
22. Bernard A, Boumsell L. The clusters of differentiation (CD) de- ischaemic heart disease. Drugs 1998;56:629‐665.
fined by the First International Workshop on Human Leucocyte 43. Kontermann RE. Strategies to extend plasma half-lives of recombi-
Differentiation Antigens. Hum Immunol. 1984;11:1-10. nant antibodies. BioDrugs. 2009;23(93):109.
23. Fröland S, Natvig JB, Berdal P. Surface- bound immunoglobu- 4 4. Wayne AS, Fitzgerald DJ, Kreitman RJ, Pastan I. Immunotoxins for
lin as a marker of B lymphocytes in man. Nat New Biol. 1971 Dec leukemia. Blood 2014;123:2470‐2477.
22;234(51):251-252. 45. Kaplon H, Reichert JM. Antibodies to watch in 2019. MAbs.
24. Guglielmi P, Preud'homme JL. Immunoglobulin expression in human 2019;11:219‐238.
lymphoblastoid cell lines with early B cell features. Scand J Immunol. 46. Nisonoff A, Rivers MM. Recombination of a mixture of univalent
1981;13:303‐311. antibody fragments of different specificity. Arch Biochem Biophys.
25. Miller JF. Role of the cells which originate from the thymus and 1961;93:460‐462.
bone marrow. Ann Immunol (Paris). 1974;125C:213-229. 47. Brinkmann U, Kontermann RE. The making of bispecific antibodies.
26. Engel P, Boumsell L, Balderas R, et al. CD Nomenclature 2015: MAbs. 2017;9:182‐212.
human Leukocyte Differentiation Antigen Workshops as a Driving 48. Davico Bonino L, De Monte LB, Spagnoli GC, et al. Bispecific mono-
Force in Immunology. J Immunol. 2015;195:4555-4563. clonal antibody anti-CD3 x anti-tenascin: an immunotherapeutic
27. Zola H, Swart B. The human leucocyte differentiation antigens agent for human glioma. Int J Cancer 1995;61:509‐515.
(HLDA) workshops: the evolving role of antibodies in research, di- 49. Smits NC, Sentman CL. Bispecific T-Cell engagers (BiTEs) as treat-
agnosis and therapy. Cell Res. 2005;15:691-698. ment of B-Cell lymphoma. J Clin Oncol 2016;34:1131‐1133.
28. Reinherz EL, Kung PC, Goldstein G, Schlossman SF. Further charac- 50. Levine BL, Miskin J, Wonnacott K, Keir C. Global Manufacturing of
terization of the human inducer T cell subset defined by monoclonal CAR T Cell Therapy. Mol Ther Methods Clin Dev 2017;4:92‐101.
antibody. J Immunol. 1979;123:2894‐2896. 51. Jones TD, Carter PJ, Plückthun A, et al. The INNs and outs of anti-
29. Nolan GP. Flow cytometry in the post fluorescence era. Best Pract body nonproprietary names. MAbs. 2016;8:1.9
Res Clin Haematol. 2011;24:505-508. 52. Mallbris L, Davies J, Glasebrook A, Tang Y, Glaesner W, Nickoloff
3 0. Tonegawa S. Somatic generation of antibody diversity. Nature. BJ. Molecular insights into fully human and humanized monoclo-
1983;302:575-581. nal antibodies: what are the differences and should dermatologists
31. Lefranc MP. Antibody Informatics: IMGT, the International care? J Clin Aesthet Dermatol. 2016;9:13‐15.
ImMunoGeneTics Information System. Microbiol Spectr. 2014;Apr
2(2).
32. Davis MM. Molecular genetics of the T cell-receptor beta chain.
How to cite this article: Béné MC. The wonderful story of
Annu Rev Immunol. 1985;3(537):560.
monoclonal antibodies. Int J Lab Hematol.
33. Lalonde ME, Durocher Y. Therapeutic glycoprotein production in
mammalian cells. J Biotechnol. 2017;251:128‐240. 2019;41(Suppl. 1):8‐14. https://doi.org/10.1111/ijlh.13006
3 4. Dos Santos ML, Quintilio W, Manieri TM, Tsuruta LR, MoroBraz
AM. Advances and challenges in therapeutic monoclonal antibodies
drug development. Braz J Pharm Sci. 2018;54(Special):e01007.