search
for
 About Bioline  All Journals  Testimonials  Membership  News


Biotecnologia Aplicada
Elfos Scientiae
ISSN: 0684-4551
Vol. 12, Num. 2, 1995, pp. 115-119
Biotecnologia Aplicada 12(2): 115-119

ARTICULO SOBRE TECNICAS / PAPER ON TECHNIQUE

SENDAI VIRUS REMOVAL AND INACTIVATION DURING MONOCLONAL ANTIBODY PURIFICACION.

Rodolfo Valdes^1, Tania Diaz^1, Aymara Nieto^2, Calixto Garcia^2, Maite Perez^1, Janet Garcia^1,Yair Quinones^3

1Monoclonal Antibody Production Department. Center for Genetic Engineering and Biotechnology.P.O.Box 6162, Havana 6, Cuba.^ 2Quality Control Department of the National Center for Laboratory Animal Production. P.O.Box 6162 Havana 6, Cuba. ^3Hepatitis B Vaccine Department. Center for Genetic Engineering and Biotechnology. P.O.Box 6162. Havana 6, Cuba

Code Number: BA95048
File Sizes:
     Text: 24K
     Graphics: Line Drawings (gif) 28K

Recibido en agosto de 1994. Aprobado en febrero de 1995.

Key words: Sendai virus, monoclonal antibody, Hepatitis B, virus validation.

SUMMARY

A feature common to all biologicals in whose production any material of animal or human origin have been used is the risk of viral contamination. Validation of viral removal during their manufacture is one of several techniques which can be used to establish product safety. The usefulness of this validation technique in the manufacture of monoclonal antibody CB/HEP/1 is demonstrated in this paper. Several procedures were used to examine ways eliminating infectious viruses by experimenting with a scale-down manufacturing procedure, and spiking of individual step or the undivided purification process to determine the extent of removal and viral inactivation. In this case cumulative clearance factor of 16 logs was achieved for the Sendai virus used as a relevant and model virus. It can be concluded that this clearance factor provides a highly safe purification standard in the production process of monoclonal antibody from ascitic fluid.

RESUMEN

Un aspecto comun para todos los productos biologicos en cuya produccion se involucra el uso de algun material de origen humano o animal, es el riesgo de contaminacion viral. La validacion de la eliminacion de agentes virales durante la fabricacion de un producto es una de las tecnicas que se emplean para establecer la seguridad del mismo. La utilidad de las tecnicas de validacion puede ser ilustrada en la manufactura del anticuerpo monoclonal CB/HEP/1 dirigido contra el antigeno de superficie del virus de la Hepatitis B. Varios procedimientos se basan en el principio de la eliminacion de virus infecciosos mediante experimentos que involucran un desescalado del proceso de purificacion, desafio del proceso o de cada paso individualmente, para determinar el poder de eliminacion y de inactivacion de virus. En este caso, el factor total de eliminacion encontrado para el virus Sendai fue de 16 logs, por lo que se concluye que este proceso de purificacion brinda un elevado margen de seguridad para anticuerpos obtenidos a partir de fluido ascitico.

INTRODUCTION

Recent biotechnological developments have brought new quality demands into the pharmaceutical manufactures for human use. Many of this products are based on monoclonal antibodies or recombinant DNA technologies and most of them are purified from cell tissues, organs, blood or other biological fluids. The potential for contamination by infectious viruses comes from many sources. A major problem is the fact that cell line particular those of rodent or human origin commonly used in the production of biopharmaceuticals often contain endogenous or latent retroviruses (1 and 2).

In the past, a number of biologicals administered to humans was contaminated with viruses, and in several instances the contaminant was identified only many years after the product's introduction into the market. Yellow fever vaccine was contaminated with Avian Leukosis virus by virtue of its production in infected hen eggs and also by Hepatitis B virus contained in human sera added to the vaccine as a stabilizer. Other example is Sendai virus 40 contamination of polio virus and adenovirus vaccines prepared in the 1950s on primary cultures of kidney cells obtained from rhesus monkeys which naturally harboured an imperceptible clinical infection with Sendai virus 40, and HIV has also contaminated blood products (2).

Three main approaches can be adopted to control potential virus contamination of biologicals: selecting and testing source materials for the absence of the viruses, testing the capacity of the production processes to remove or inactivate viruses and testing the product for contaminating viruses at appropriate stages of production (2).

    Fig. 1 Blocking flow diagram downstream processing MAb CB/HEP/1

Validation studies are performed in which specific processing step are challenged with a high titer of infectious viruses. The objective of validation is to estimate quantitatively the overall level of virus reduction obtained during the various stages of purification and/or viral inactivating stages.

This will be achieved by the deliberate addition (spiking) of significant amounts of a virus to the row material to be purified and to different fractions obtained during the various purification stages. A major aim in performing a validation is the determination of which viruses should be used. These fall in two categories; relevant virus and model virus. relevant viruses are viruses which are known or likely to contaminate the source material or other materials used in the production process.

Model viruses: if the use of relevant viruses does not encompass viruses with a wide range of physico-chemical properties, then validation should be performed with model viruses. Validation of Ab CB/HEP/1 purification process was performed on a research scale (figure 1) and this monoclonal antibody purified from BALB/c ascitic fluid. The viral agent used in this study was an RNA virus, family Paramyxoviridae, genus Paramyxovirus, species parainfluenza 1[Sendai virus] (table 1).

The virus particles are spherical, 150-200 nm in diameter, have a helical nucleocapsid, a continuous single RNA genome and low resistance to physical and chemical agents such as UV light, temperatures above 37^oC, lipid solvents and extremes pHs (3). This virus provides a representative panel of possible mouse viral contaminant such as Retrovirus, Influenza or other small enveloped viruses (4).

MATERIALS AND METHODS

Purification of Monoclonal Antibodies from Ascitic Fluid

Samples of ascitic fluid were obtained from routine large scale ascitic fluid production derived from hybridome cell line CB/HEP/1, which secretes a murine monoclonal antibody (IgG2b) specific for the rHBsAg(5). The viral removal experiments were performed with ascitic fluid and inactivation experiments were performed with purified monoclonal antibody.

To design the virus study validation, a scaling down on the purification process was done. For our experiments; the scale-down was brought to the 1% of the real production scale and the level of purification of the product mimiked the production process and the products generated were similar in terms of purity, specific activity, mouse DNA and yield. (table 2).

Elisa

PVC plates were coated with rHBsAg (100 uL, 10 ug/mL in NaHCO3 100 mM, pH 9.6, 20 min at 56^oC). The plates were washed with PBS/ 0.05% Tween-20. Several dilutions of the samples were added and incubated during two hours at RT.

The plates were washed again and incubated at 37^oC for one hour with HRPO-labeled, affinity purified goat anti mouse-IgG diluted 1/1000 in PBS/Tween-20/BSA. The plates were washed and subsequently incubated 20 min with substrate solution (5 uL H2O2 and 5 mg OPD diluted in 10 mL of Citrate 50 mM, pH 5.5).

The reaction was stopped with 250 mM SO4H2 (50 uL/well) and the absorbency was measured at 492 nm in an ELISA reader. The average value curve of CB/HEP/1 was used to quantify Ab activity.

Electrophoresis

Protein purity was analyzed by electrophoresis in 12.5% polyacrylamide gel containing 0.1% SDS at pH 8.6 as described by Laemmli (6). Prior to electrophoretic run, eluates were treated with 1% SDS at 50EC for 10 min. After electrophoresis, protein in the gel was stained with Commassie brilliant blue R-250.

Protein Concentration

The protein concentration was determined according to Lowry et al.(7).

Murine DNA

The murine DNA in purified monoclonal antibody preparations was assayed by dot-blot hybridization using a mouse genomic DNA probe. Standard mouse DNA was used to determine the DNA content in the same experiment.

Table 1. Specification of the virus strain and read-out systems used for measuring the reduction and inactivaton factors of Sendai virus

--------------------------------------------------------------
Virus            Infectivity titre of     Cell       Read-out
Classification*  stock suspention**       Culture    system
                                          Method
--------------------------------------------------------------
Family:
Paramixoviridae    10^4.5 First
                   Experiment             Embryonic
Genus:
Paramyxovirus                                        HA and
Specie:            10^8.5 Second          chicken    EID50
Parainfluenza-1    Experiment             eggs
(Sendai virus)
---------------------------------------------------------------

*CIB and CENPALAB, Cuba; **Expressed in 10^log. EID50

Spike Experiments

Viral Removal

To determine the virus reduction factor in the down stream purification process of ascitic fluid, a preparation of infectious Sendai virus was added to the initial fluid in order to measure the total reduction factor of the scale-down purification process. We worked with only one virus strain to reduce the interference from other viruses in the measurement of the infectivity titers. Before and after each purification, 2 mL of the samples were taken from each test and immediately frozen at-70^oC (2).

Viral inactivation

Citrate low pH buffer was used to elute the Ab CB/HEP/1 from the Protein A-Sepharose CL4B column. In this study, we spiked the citrate buffer with Sendai virus in order to test its ability to inactivate this virus. We conducted antibody inactivating studies at 4^oC, 37^oC and 60^oC for 10 h to determine the condition in which the antibody does not lose its recognition capacity of the antigen and subsequent measure of the virus stability (table 3).

Table 2. Sendai virus reduction factor of CB/HEP/1 purification method

---------------------------------------------------------------

                          HA   EID50  HA   EID50  HA    EID50
STEPS                     I*   I      II*  II     III*  III
---------------------------------------------------------------
Ascitic Fluid            128   4.2    64    4.4   8192  8.4
Precipitation I          8     2.2    8     2.2   6.4   3.4
Precipitation II         8     1.9    8     1.9   8     2
Precipitation Pellet II  256   3.9    256   3.9   8192  6.2
Desalted Fraction        256   3.3    32    3.3   8192  7.2
AC Non bound (1)         256   4.2    64    4.2   1638  7.2
AC Eluted (1)            4     0      4     0     4     0
AC Non bound (2)         256   4.4    64    4.4   1638  7.2
AC Eluted (2)            4     0      8     0     2     0
Gel Filtration (1)       2     0      4     0     2     0
Gel Filtration (2)       -     0      0     0     2     0
100mM Citric acid pH3    4     0      4     0     0     0
150mM PBS pH 8           -     0      0     0     0     0
20mM Tris, 150mM
NaCl pH7.6               2     0      2     0     0     0
Specific Activity [%]    136   -      133   -     90    -
Purity [%]               92    -      96    -     91    -
Murine DNA (pg/mgAb)    <10    -     <10    -    <10  
 -
Yield mgAb/MLAsc         1.6   -      1.7   -     1.5   -
---------------------------------------------------------------

*Hemagglutination titre expressed as the reciprocal of the dilution still showing HA. AC. Protein A affinity chromatography.

Hemagglutination Test

An HA test was carried out to determine the infective titers of Sendai virus. For this purpose two log serial dilutions were made in PBS 7.4 and human erytrocyte sedimentation was observed after 30-45 min at room temperature (22^oC). The titers were expressed as the reciprocal of the dilution still showing hemagglutination.

Embryonated Egg Infectivity Doses

The Reed-Muech method was applied to determine the infectivity titers. Several dilutions of the samples were made in PBS and 0.1 mL of these dilutions were inoculated in the allaontoic cavity of embryonated chicken eggs (8).

Clearance/Inactivation Factor

The virus reduction factor of a purification or inactivation step is defined as the log10 of the ratio between the virus load in the preparation material and the virus load in the post-purification material which is ready for the next step of the process. The following abbreviations are used ( 9):

Starting material: vol v'; titer 10a'

Viral load: v'*10a'

Final material: vol v"; titer 10a" Viral load: v"*10a"

The individual reduction (RF) or inactivation factor (IF) is calculated according to 10rf = v'*10a'/ v"*10a"

RESULTS AND DISCUSSION

Sendai virus inactivation

A low pH is frequently used to elute proteins from affinity chromatography. In our purification process we used 0.1 M, pH 3 citrate buffer to elute CB/HEP/1 from Protein A Sepharose CL-4B column (Pharmacia) (5). Heat treatment however, has a mayor disadvantage: not all products tolerate heat and this could have an adverse effect on the efficacy, stability or biochemistry of the product (10). Kinetic studies for inactivation of CB/HEP/1 have been important in demonstrating at which condition this monoclonal antibody was more resistant. For example, CB/HEP/1 Ab were completely inactivated after one hour at 60^oC in citrate buffer pH 3 while after 10 h at 37^oC the activity was 23.4%. For this reason, we selected citrate pH 3 at 4^oC as the best experimental condition as in which, after seven h the activity was more than 80% of the initial activity (table 3).

This buffer was spiked with Sendai virus. We conducted the inactivation studies at 4^oC and 7 h for periods from one hour to 7 h. The reduction in Sendai virus titers was 4.2 logs (first experiment) and 8.4 logs (second experiment) (tables 4 and 5). Inactivation of this virus occured rapidly, and was dependent on buffer formulation and not on temperature because the reduction in virus titer used as control (PBS, pH 7.4 at 4^oC) was not observed (table 4).

Table 3. Ability of citric acid pH3 treatment at different temperatures to inactivate a monoclonal antibody CB/HEP/1 preparation

----------------------------------------------------------------

         4 C             37 C         60 C           4 C
     Ab   Activity   Ab   Activity Ab   Activity  AB   Activity
Time conc I          conc I        conc I         conc I
[h]  I    %          I    %        I    %         I    %
----------------------------------------------------------------
0    11.5 100.0      11.5 100.0    11.5 100.0     12.5  100.0
1/2  11.2 97.3       8.6  74.7     0.4  3.4       12    96.3
1    12.5 108        8.3  80.0     0.0  0.0       12.1  96.7
2    11.1 96.5       7.6  66.0     0.0  0.0       12.5  100.0
3    11.8 102        7.0  60.8     0.0  0.0       12.1  96.7
4    10.5 91.3       6.6  57.3     0.0  0.0       11.9  95.2
5    10.2 88.6       6.5  56.7     0.0  0.0       7.9   63.6
6    10.2 88.6       6.3  57.4     0.0  0.0       7.7   61.9
7    10.1 87.8       6.0  52.1     0.0  0.0       6.9   55.2
8    9.4  81.7       4.5  39.1     0.0  0.0       6.5   52.0
9    8.4  73.0       3.3  28.6     0.0  0.0       6.3   50.2
10   7.7  66.9       2.7  23.4     0.0  0.0       6.3   50.2
----------------------------------------------------------------

I first experiment, II second experiment, Activity (FC/IC)*100.0

FC final concentration measurement by ELISA (mg/ml)

IC initial concentration measurement by ELISA (mg/ml)

Sendai viral removal

Validation was performed on a research scale that represented 1% of the real scale of the purification process (figure 1) (11). The Ab purity, especific activity and yield provided by the protocol are equivalent to real production scale, 50 mL of ascitic fluid were spiked with Sendai virus. This virus is representative of a panel of possible contaminating agents with similar physico-chemical structures: a murine retrovirus and other viruses of RNA, envelope, size between 100-250 nm, spherical shape and little resistance to physico-chemical agents.

The removal of the viral agent in this standard purification system worked only on protein A affinity chromatography. There is a reduction factor of up to 8.4 logs (table 2). The overall clearance factor (inactivating and removal factor, 16.8 logs) was substantially greater than the potential virus titer in the source material; concentration of this group of contaminating viruses in a cell culture rarely exceeds 10^6 (11) and ascitic fluid 10^9 (12). Now we are making spiking experiments of the purification process with other viral particles that represent a different viral system in order to complete the panel representing a model virus group in terms of physico-chemical properties of all murine viruses.

Table 4. Ability of citric acid pH 3 at 4 C treatment to inactivate the Sendai virus.

--------------------------------------------------------------
            EID   Ab   Act   EID    Ab   Act  EID   Ab   Act
Time Vol    50   conc   %    50    conc   %   50    conc  %
[h]  [ml]   I     I     I    II     II    II  III   III  III
---------------------------------------------------------------
0     1   10^4.0 0.99  100.0 10^4.4 0.99 100.0 104.2 0.99 100.0
1     1   10^0   0.80  80.8  10^0   ND   ND    10^0  1.08 109
2     1   10^0   0.78  78.7  100    1.18 118   10^0  1.7  171
3     1   10^0   1.60  160   100    0.6  55.9  10^0  1.16 117
4     1   10^0   1.40  140   100    0.95 95.9  10^0  0.81 80.8
5     1   10^0   1.20  120   100    0.61 61.6  10^0  1.26 129
6     1   10^0   1.56  156   100    0.51 51.5  10^0  1.06 107
7     1   10^0   0.80  80.8  100    0.73 73.7  10^0  0.91 91.9
AcC
pH7   1   10^0    -     -    100     -    -    10^0   -    -
----------------------------------------------------------------

I: first experiment II: second experiment III:third experiment AcAph7: citric acid pH7 ND: not determined conc: Concentration measurement by ELISA.

Table 5. Ability of citric acid pH 3 at 4 C treatment to inactivate the Sendai virus


-------------------------------------------------------
Time(h)  Vol(ml)  EID50  IE  DE  mg Ab/ml   Act(%)
------------------------------------------------------- 
0         1      10^8.4  15   3    1.53      100.0
1         1      10^0    15   0    1.01      66
2         1      10^0    15   2    1.37      90
3         1      10^0    15   0    1.15      75
4         1      10^0    15   1    1.28      83
5         1      10^0    15   0    1.45      94.7
6         1      10^0    15   1    1.82      119
7         1      10^0    15   2    1.94      126
AcC pH 7  1      10^0    15   2     -        -

--------------------------------------------------------

AcA: citric acid IE: infectivated eggs DE: dead eggs

A cumulative removal of more than 16 logs of removal/inactivation can be estimated if it is assumed that the steps are independent events. The degree of removal shown by these logs would be more reliable if the actual amount of virus contaminating the ascitis bulk were known.

We are trying at present to measure the virus level in ascitic fluid risk in CB/HEP/1 in order to determine the real concentrations of the different viruses that are included in this group, and as well as we are using a MAP Hageman test on the master cell bank, ascitic fluid and final product as an additional test to confirm the safety of the manufacturing process. In all cases, the results were negative for this kind of virus, suggesting that there is a substantial margin of safety in the process and for Hepatitis B vaccine since this monoclonal antibody is used to immunopurify the recombinat Hepatitis B surface antigen used in this biological preparation.

ACKNOWLEDGMENTS

Our thanks to Lic. Beatriz Gago, Dr. Jorge Gavilon-do, Lic. Suany Ojeda and Ing. Alberto Agrass for critical review of this manuscript and Dr. Luis Herrera for his scientific contributions.

REFERENCES

1. EXPERT COMMITTEE ON BIOLOGICAL STANDARIZATION, (1991). Requirements for Biological Sustances. In: Proposed requirements for monoclonal antibodies for clinical use in humans. 48:1-23

2. WHITE, E.; J. B. GRUN; SUR. SON-CHONG; F. A. SITO, (1991). Process validation for virus removal and inactivation. Biopharm, April-May; 34-37.

3. PARKER, J. C; C. B. RICHTER, (1982). Viral deseases of the respiratory system. The Mouse in Biomedical Research II:109-158.

4. Committe for proprietary medical products ad hoc working party on biotechnology/pharmacy and working party on safety on medicine (1991) EEC regulatory document mate for guidance validation. Validation of virus removal and inactivation. Biologicals 19:247-251.

5. FONTIRROCHI, G.; M. DUENAS; E. FERNANDEZ; P. FUENTES; M. PEREZ; D. MAINET; M. AYALA; J. GAVILONDO; C. DUARTE(1993). A mouse hybridoma cell line secreting IgG and IgM with specificity for the Hepatitis B Surface Antigen Virus Requirements for Biological Sustances. Biotecnologia Aplicada 10:25-30

6. LAEMMLI. U. K, (1970). Cleavage of structural proteins during the assembly by bacteriophage T 4. Nature 270:680-685.

7. LOWRY, O. H.; N. J. ROSEMBROUGH; A. L FARR and R. J. RANDAL, (1951) Protein measurements with folin-phenol reagent. J. Biol Chemical 193:256-269.

8. REED. L.; J and H. A. MUECH, (1938). A simple method of stemativy fifty per cent endpoints. Amer. J. Hyg 27:493-487.

9. THOMAS. C.; HAGEMAN, T.C. (1991). A analysis of clearance factor measurements performed by spiking experiment. Biopharm, July-Agust; 39-41.

10. GRUN J. B.; E. WHITE and F. A. SITO (1992).Viral Removal and Inactivation by purification on biopharmaceuticals: Biopharm, November-December; 22-26.

11. MARIANI. M and L. ARDITI (1992).Validating the Preparation of Clinical Monoclonal Antibodies. Biotechnology 10:394-396.

12. AVEST, R. A.; J. J. ZOULEN; E. M. SPIJKERSEM; A. OSTERHAUS; G. STEENIS; C. KREYL (1992). Purification Process Monitoring in Monoclonal Antibody Preparation: Contamination with Viruses, DNA and Peptide Growth Factor. Biologicals 20:177-186


The following images related to this document are available:

Line drawing images

[ba95048a.gif]
Home Faq Resources Email Bioline
© Bioline International, 1989 - 2024, Site last up-dated on 01-Sep-2022.
Site created and maintained by the Reference Center on Environmental Information, CRIA, Brazil
System hosted by the Google Cloud Platform, GCP, Brazil